Language selection

Search

Patent 3014033 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 3014033
(54) English Title: COMPOUNDS, COMPOSITIONS, AND METHODS FOR THE TREATMENT OF INFLAMMATORY, DEGENERATIVE, AND NEURODEGENERATIVE DISEASES
(54) French Title: COMPOSES, COMPOSITIONS, ET PROCEDES DE TRAITEMENT DE MALADIES INFLAMMATOIRES, DEGENERATIVES, ET NEURODEGENERATIVES
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • C07C 51/09 (2006.01)
(72) Inventors :
  • BAZAN, NICOLAS G. (United States of America)
  • PETASIS, NICOS A. (United States of America)
(73) Owners :
  • UNIVERSITY OF SOUTHERN CALIFORNIA (United States of America)
  • BOARD OF SUPERVISORS OF LOUISIANA STATE UNIVERSITY AND AGRICULTURAL AND MECHANICAL COLLEGE (United States of America)
(71) Applicants :
  • UNIVERSITY OF SOUTHERN CALIFORNIA (United States of America)
  • BOARD OF SUPERVISORS OF LOUISIANA STATE UNIVERSITY AND AGRICULTURAL AND MECHANICAL COLLEGE (United States of America)
(74) Agent: MARKS & CLERK
(74) Associate agent:
(45) Issued: 2024-02-27
(86) PCT Filing Date: 2016-02-09
(87) Open to Public Inspection: 2016-08-18
Examination requested: 2021-01-14
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2016/017112
(87) International Publication Number: WO2016/130522
(85) National Entry: 2018-08-08

(30) Application Priority Data:
Application No. Country/Territory Date
62/113,893 United States of America 2015-02-09
62/120,229 United States of America 2015-02-24

Abstracts

English Abstract

This disclosure provides compounds, pharmaceutical compositions, and methods of use for the prevention and treatment of inflammatory diseases or degenerative diseases including neurodegenerative diseases. Embodiments of the present disclosure provide compounds related to very long chain polyunsaturated fatty acids, pharmaceutical compositions containing the provided compounds, and methods of treating a subject with a condition or disease using provided compounds or pharmaceutical compositions.


French Abstract

La présente invention concerne des composés, des compositions pharmaceutiques, et des procédés d'utilisation pour la prévention et le traitement de maladies inflammatoires ou de maladies dégénératives comprenant des maladies neurodégénératives. Les modes de réalisation de l'actuelle présentation prévoient des composés liés à des acides gras polyinsaturés à très longue chaîne, des compositions pharmaceutiques contenant les composés prévus, et des procédés de traitement d'un sujet présentant un état ou affecté d'une maladie en utilisant les composés ou les compositions pharmaceutiques prévus.

Claims

Note: Claims are shown in the official language in which they were submitted.


What is claimed:
1. A compound having the general structure G or H,
Image
wherein:
n is 0 to 19; and
the R group is methyl, ethyl, alkyl, or a cation forming a pharmaceutically
acceptable
salt which is: ammonium cation, iminium cation, or a metal cation.
2. The compound of Claim 1, wherein n is 0 to 13.
3. The compound of Claim 1, wherein n is 1, 3, 5, 7, 9, 11 or 13.
4. The compound of Claim 1, wherein n is 0, 2, 4, 6, 8, 10 or 12.
5. The compound of Claim 1, wherein n is 9 or 11.
6. The compound of Claim 1, wherein the R group is methyl or ethyl.
7. The compound of Claim 1, wherein the R group is the metal cation which is
sodium,
potassium, magnesium, zinc, or calcium cation.
8. The compound of Claim 1, wherein the compound is l, J, K, or L, having
one of the
following structures:
46

Image
wherein:
n is 0 to 19; and
the R group is methyl, ethyl, alkyl, or a cation forming a pharmaceutically
acceptable
salt which is: ammonium cation, iminium cation, or the metal cation.
9. The compound of Claim 1, wherein the compound is (S, 16Z, 19Z, 22Z, 25Z,
27E, 31Z)-
29-hydroxytetratriaconta-16, 19,22,25,27,31-hexaenoic acid that has the
following structure:
47

Image
wherein R is methyl, ethyl, alkyl, or a cation forming a pharmaceutically
acceptable salt
which is: ammonium cation, iminium cation, or a metal cation.
10. A compound having the general structure M or N,
Image
wherein:
n is 0 to 19; and
the R group is hydrogen, methyl, ethyl, alkyl, or a cation forming a
pharmaceutically
acceptable salt which is: ammonium cation, iminium cation, or a metal cation.
11. The compound of Claim 10, wherein n 0 to 13.
12. The compound of Claim 10, wherein n 1, 3, 5, 7, 9, 11 or 13.
13. The compound of Claim 10, wherein n is 0, 2, 4, 6, 8, 10 or 12.
48

14. The compound of Claim 10, wherein n is 9 or 11.
15. The compound of Claim 10, wherein the R group is methyl or ethyl.
16. The compound of Claim 10, wherein the R group is the metal cation which is
sodium,
potassium, magnesium, zinc, or calcium cation.
17. The compound of Claim 10, wherein the compound is 0, P, Q, R, S, T, U or V
having
one of the following structures:
49

Image

wherein:
n is 0 to 19; and
the R group is hydrogen, methyl, ethyl, alkyl, or a cation forming a
pharmaceutically
acceptable salt which is: ammonium cation, iminium cation, or the metal
cation.
18. The compound of Claim 10, wherein the compound is: (14Z,
17Z,20R,21E,23E,25Z,275,29Z)-20,27-dihydroxydotriaconta-
14,17,21,23,25,29-hexaenoic acid; sodium (14Z,17Z,20R,21E,23E, 25Z,27S,29Z)-
20,27-
dihydroxydotriaconta-14,17,21,23,25,29-hexaenoate; methyl (14Z,17Z,20R,21
E,23E,25Z,27S,29Z)-20,27-dihydroxydotriaconta-14, 17,21,23,25,29-hexaenoate;
(16Z,
19Z,22R,23E,25E,27Z,29S,31Z)-22,29-dihydroxytetratriaconta-16,19,23,25,27,31-
hexaenoic acid; sodium (16Z,19Z,22R,23E,25E,27Z,29S,31Z)-22,29-
dihydroxytetratriaconta-16,19,23,25,27,31-hexaenoate; or methyl (16Z,
19Z,22R,23E,25E,27Z,29S,31Z)-22,29- dihydroxy-tetratriaconta-16,
19,23,25,27,31-
hexaenoate, which have the following structures:
Image
51

19. A compound having the general structure X or Z:
Image
wherein:
n is 0 to19; and
the R group is hydrogen, methyl, ethyl, alkyl, or a cation forming a
pharmaceutically
acceptable salt which is an ammonium cation, iminium cation, or a metal
cation.
20. The compound of Claim 19, wherein n is 0 to 13.
21. The compound of Claim 19, wherein n is 1, 3, 5, 7, 9, 11 or 13.
22. The compound of Claim 19, wherein n is 0, 2, 4, 6, 8, 10 or 12.
23. The compound of Claim 19, wherein n is 9 or 11.
24. The compound of Claim 19, wherein the R group is methyl or ethyl.
25. The compound of Claim 19, wherein the R group is hydrogen.
26. The compound of Claim 19, wherein the compound is any one of the following

structures, wherein R is hydrogen, methyl or sodium:
52

Image
27. A pharmaceutical composition comprising a compound according to any one of
claims
1-26 and a pharmaceutically acceptable carrier.
28. Use of an effective amount of a compound according to any one of claims 1-
26 and a
pharmaceutically acceptable carrier for treating an inflammatory disease,
degenerative
disease or neurodegenerative disease in a subject in need thereof.
29. Use of an effective amount of a compound according to any one of claims 1-
26 and a
pharmaceutically acceptable carrier for the manufacture of a medicament for
treating an
inflammatory disease, degenerative disease or neurodegenerative disease in a
subject in
need thereof.
30. The use according to Claim 28 or 29, wherein the disease is: rheumatoid
arthritis,
osteoarthritis, atherosclerosis, cancer, diabetes, intestinal bowel disease,
prostatitis,
multiple sclerosis, autism, schizophrenia, depression, traumatic brain injury,
status
epilepticus, Huntington's disease, Alzheimer's disease, Parkinson's disease,
amyotrophic
lateral sclerosis, retina degenerative disease, age-related macular
degeneration, inherited
retinal degenerative disease, Stargardt-like macular dystrophy, X-linked
juvenile
retinoschisis, perioperative hypoxia, retinitis pigmentosa or glaucoma.
31. The use according to Claim 28 or 29, wherein the disease is ischemic
stroke.
53

32. Use of a pharmaceutical composition according to Claim 27 for treating an
inflammatory disease, degenerative disease or neurodegenerative disease in a
subject in
need thereof.
33. Use of a pharmaceutical composition according to Claim 27 for the
manufacture of a
medicament for treating an inflammatory disease, degenerative disease or
neurodegenerative disease in a subject in need thereof.
34. The use according to Claim 32 or 33, wherein the disease is: rheumatoid
arthritis,
osteoarthritis, atherosclerosis, cancer, diabetes, intestinal bowel disease,
prostatitis,
multiple sclerosis, autism, schizophrenia, depression, traumatic brain injury,
status
epilepticus, Huntington's disease, Alzheimer's disease, Parkinson's disease,
amyotrophic
lateral sclerosis, retina degenerative disease, age-related macular
degeneration, inherited
retinal degenerative disease, Stargardt-like macular dystrophy, X-linked
juvenile
retinoschisis, perioperative hypoxia, retinitis pigmentosa or glaucoma.
35. The use according to Claim 32 or 33, wherein the disease is ischemic
stroke.
36. Use of a therapeutically effective amount of a compound of any one of
claims 1 to 26
for treating a condition or a disease in a subject suffering from the
condition or the disease.
37. Use of a therapeutically effective amount of a compound of any one of
claims 1 to 26
for the manufacture of a medicament for treating a condition or a disease in a
subject
suffering from the condition or the disease.
38. The use of Claim 36 or 37, wherein the disease is an inflammatory disease,
a
degenerative disease, a neurodegenerative disease, or an eye disease.
39. A use of Claim 36 or 37, wherein the disease is retinitis pigmentosa, age-
related
macular degeneration, Stargardt-like macular dystrophy, or X-linked juvenile
retinoschisis.
40. Use of a compound according to any of one Claims 1-26 for increasing the
expression
of SIRT1 in a subject.
54

41. Use of a compound according to any of one Claims 1-26 for the manufacture
of a
medicament for increasing the expression of SIRT1 in a subject.

Description

Note: Descriptions are shown in the official language in which they were submitted.


COMPOUNDS, COMPOSITIONS, AND METHODS FOR THE TREATMENT OF
INFLAMMATORY, DEGENERATIVE, AND NEURODEGENERATIVE DISEASES
CROSS REFERENCE TO RELATED APPLICATIONS
This application claims the benefit of U.S. Provisional Application No.
62/113,893,
filed February 9, 2015, and of U.S. Provisional Application No. 62/120,229,
filed February
24, 2015.
This invention was made with government support under RO1 EY005121 and P30
GM103340 awarded by the National Institutes of Health. The government has
certain rights
in the invention.
FIELD OF THE DISCLOSURE
This disclosure relates to previously unknown compounds, compositions, and
methods of use for the prevention and treatment of inflammatory, degenerative,
and
neurodegenerative diseases, including related conditions or disorders. More
specifically, this
disclosure relates to compounds related to very-long-chain polyunsaturated
fatty acids and
their hydroxylated derivatives, and their use as therapeutic agents. In
particular, this
disclosure relates to the treatment of stroke and other brain conditions
involving
neurodegeneration, as well as retinal degenerative diseases and related
conditions causing
loss of sight or blindness.
BACKGROUND OF THE DISCLOSURE
Inflammatory, degenerative, and neurodegenerative diseases include a large
number
of diseases that affect a very large number of people worldwide. In most
cases, these
diseases and related conditions and disorders are difficult to treat, and
remain as an unmet
medical need.
Inflammatory diseases in the scope of this disclosure include acute and
chronic
disorders were homeostasis is disrupted by abnormal or dysregulated
inflammatory
response. These conditions are initiated and mediated by a number of
inflammatory factors,
including oxidative stress, chemokines, cytokines, breakage of blood/tissue
barriers,
autoimmune diseases, genetic factors being gene susceptibility, polymorphisms
or inherited
conditions, or other conditions that engage leukocytes, monocytes/macrophages
or
parenchymal cells that induce excessive amounts of pro-cell injury, pro-
inflammatory/disruptors of cellular and /or organ homeostasis. These diseases
occur in a
wide range of tissues and organs and are currently treated, by anti-
inflammatory agents
such as corticosteroids, non-steroidal anti-inflammatory drugs, TNF
modulators, COX-2
inhibitors, etc.
1
Date Recue/Date Received 2022-07-22

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Degenerative diseases comprise conditions that involve progressive loss of
vital cells
and tissues that result in progressive impairment of function, such as loss of
cartilage in
knees, hip joints or other joints such as in osteoarthritis. Other
degenerative diseases
engages cellular and intercellular homeostasis perturbations and includes
heart disease,
.. atherosclerosis, cancer, diabetes, intestinal bowel disease, osteoporosis,
prostatitis,
rheumatoid arthritis, etc.
Neurodegenerative diseases include some of the major diseases of the brain,
retina,
spinal cord and peripheral nerves, whereby a failure upon neuroinflammatory
induction leads
to a progressive demise of cellular organization including neuronal onset cell
death leading
.. to impaired function. These are due to immune or inflammatory disorders
and/or inherited
conditions or age-related pathologies. They include ischemic stroke,
Alzheimer's disease,
Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis,
autism, neuropathic
pain, traumatic brain injury, schizophrenia, depression, and retinal
degenerative diseases
such as age-related macular degeneration, glaucoma, inherited eye diseases
such as
.. retinitis pigmentosa, Stargardt disease, Stargardt-like macular dystrophy,
etc.
Retinal degenerative diseases are the leading causes of blindness. Retinal
degeneration is the deterioration of the retina caused by the progressive and
eventual death
of the photoreceptor and retinal pigment epithelial cells of the retina.
Retinitis pigmentosa
affects between 50,000 and 100,000 people in the United States alone, and
macular
degeneration is the leading cause of vision loss for those aged 55 and older
in the United
States, affecting more than 10 million people. There are no effective
treatments for these
and other retinal degenerative diseases.
Despite progress made in understanding the pathophysiology of inflammatory,
degenerative, and neurodegenerative diseases, their detailed molecular
mechanisms remain
to be fully elucidated. Available treatments today are not able to effectively
treat these major
diseases or to slow-down their onset and progressive impairment of vital
functions. For
example, in the case of retinal degenerative diseases, the detailed processes
involved in the
progressive loss of photoreceptor cells remain unknown, and available
treatments today are
not able to effectively treat these major diseases and prevent loss of sight.
Therefore, there is a major therapeutic void for the prevention, treatment,
and overall
management of inflammatory, neuroinflammatory, degenerative and
neurodegenerative
diseases.
BRIEF SUMMARY OF THE DISCLOSURE
This disclosure provides compounds, compositions, and methods of use for the
treatment of inflammatory, degenerative, and neurodegenerative diseases.
The provided compounds are related to omega-3 very long chain polyunsaturated
fatty acids (n3 VLC-PUFA or VLC-PUFA) with carbon chains containing from 23 to
42
2

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
carbons. In some embodiments the provided compounds are chemically modified
pharmaceutically acceptable derivatives of VLC-PUFA containing carbon chains
from 24 to
36 carbons and include 6 or 5 alternating cis-carbon-carbon double bonds
starting at
positions 0)-3 (omega-3), co-6, co-9, cD-12, co-15 and 0)-18. In other
embodiments the
.. disclosure provides compounds that are hydroxylated derivatives of VLC-PUFA
that contain
one to two hydroxyl groups.
While VLC-PUFA containing an even number of carbons ranging from 24 to 42
carbons have been detected in the form of free acids or as components of
cellular lipids in
mammalian tissues, VLC-PUFA containing an odd number of carbons are not known
to exist
.. in nature.
The provided compounds are chemically modified pharmaceutically acceptable
derivatives to enhance their chemical and biological stability, and to enable
their use in
therapeutic applications involving various forms of drug delivery.
The provided compositions are intended for pharmaceutical use and contain
various
forms of the provided compounds and their pharmaceutically acceptable
derivatives, such as
the free carboxylic acids or their pharmaceutically acceptable salts, or as
their corresponding
esters, phospholipid derivatives, or other prodrug derivatives. The
compositions also include
additional components and formulations to facilitate solubility,
bioavailability, and stability.
The provided methods of treatment involve the use of the compounds or
compositions of the disclosure, which contain a therapeutically effective
amount of a
provided compound in one of several chemically modified forms, including the
free
carboxylic acids or their pharmaceutically acceptable salts, or as their
corresponding esters,
their phospholipid derivatives, or other prodrug derivatives.
The provided compounds, compositions, and methods of use are able to restore
.. homeostasis and induce survival signaling in certain cells undergoing
oxidative stress or
other homeostatic disruptions. Administration of a pharmaceutical composition
containing a
provided compound restores the homeostatic cellular balance and promotes the
survival of
certain cells that are essential for maintaining normal function. The provided
compounds,
compositions, and methods of use can be utilized for the preventive and
therapeutic
treatment of inflammatory, degenerative, and neurodegenerative diseases. This
provided
methods of use target critical steps of the initiation and early progression
of these conditions
by mimicking the specific biology of intrinsic cellular/organs responses to
attain potency,
selectivity, devoid of side effects and sustained bioactivity.
In particular, this disclosure provides compounds, compositions, and methods
of use
.. for the prevention and treatment of retinal degenerative diseases. The
provided compounds
and compositions induce the survival of photoreceptors and retinal pigment
epithelial cells
and protect the retina and the brain. The provided methods involve the use of
compounds
3

that induce survival signaling in both the retinal pigment epithelial cells
and photoreceptors
as well as in brain cells. Moreover this disclosure provides compounds,
compositions, and
methods of use for the prevention and treatment of stroke and other
neurodegenerative
diseases, such as epilepsy, traumatic brain damage, and spinal cord injury.
BRIEF DESCRIPTION OF THE DRAWINGS
Further aspects of the present disclosure will be readily appreciated upon
review of
the detailed description of its various embodiments, described below, when
taken in
conjunction with the accompanying drawings.
Figure 1. Postulated biosynthesis of selected hydroxylated derivatives derived
from
.. DHA (C22:6n3, or C22:6) and from omega-3 very long chain polyunsaturated
fatty acids (n3-
VLC-PUFA). Lipoxygenation of DHA with 15-LOX affords 17-HpDHA, which is either

reduced to 17-HDHA or is transformed into NPD1. DHA is also a converted by the
elongase
enzyme ELOVL4 (elongation of very long fatty acids-4) into n3-VLC-PUFA, such
as C32:6
and 034:6, which can be incorporated into phospholipids. Lipoxygenation of n3-
VLC-PUFA
with 15-LOX leads to the formation of monohydroxy derivatives of n3-VLC-PUFA,
such as
29-hydroxy 34:6 and 22,29-dihydroxy 34:6. Given the key role of the ELOVL4
enzyme, the
name "Elovanoid" is being used herein for a bioactive enzymatically-derived
hydroxylated
derivative of VLC-PUFA. Thus, the name Elovanoid 1 (ELV1) is being used for
the
20R,27S-dihydroxy 32:6 derivative, and the name Elovanoid 2 (ELV2) is being
used for the
22R,29S-dihydroxy 32:6 derivative. The stereochemistry of the mono- and di-
hydroxy
compounds were presumed to be the same as those derived from DHA.
Figure 2. Evidence of cell-derived hydroxylated derivatives (29-hydroxy-34:6
and
22,29-dihydroxy-34:6) derived from the omega-3 VLC-PUFA 34:6n3 (C34:6) from
human
retinal pigment epithelial cells in culture. Human retinal pigment epithelial
cells
(spontaneously transformed ARPE-19 cells) or primary human retinal pigment
epithelial cells
(HRPE) were incubated with 34:6n3 (100nM) during 12-16 hours and then the
culture media
collected ,lipid extracted and run in LC-MS/MS. The results suggest that C34:6
with an rin/z
of 495.5 (Fig.2A) yielded a hydroxylated product analogous to the mono-
hydroxylated DHA
derivative 17-H DHA having a parent-H m/z of 511.8 and a fragment m/z of 413,
which is
consistent with the mono-hydroxylated compound 29-hydroxy-34:6 (Fig.2B). The
data show
that compound C34:6 was also converted to an elongation product analogous to
the di-
hydroxylated DHA derivative NPD1,(10R,17S-dihydroxy-docosa-
4Z,7Z,11E,13E,15Z,19Z-
hexaenoic acid) having a parent-H m/z of 527.8, and a fragment m/z of 206
consistent with
the NPD1-like di-hydroxylated compound 22,29-dihydroxy-34:6 (Fig.2C).
Figure 3. Structures of compounds: (A) DHA, (B) NPD1, (C) C32:6, (D) C34:6,
(E)
chemically synthesized ELV1-Na, (F) chemically synthesized ELV1-Me, (G)
chemically
synthesized ELV2-Na, (H) chemically synthesized ELV2-Me.
4
Date Recue/Date Received 2022-07-22

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Figure 4. (A) Cytoprotective effects of NPD1 like compounds on OS induced
apoptosis. The results shown in this figure compare the cytoprotective
capacities of long
chain polyunsaturated fatty acids (VLC-PUFA), elovanoids ELV1 and ELV2, and
neuroprotectin D1 (NPD1) in human retinal pigment epithelial (RPE) cells
deficient in 15-
LOX-D1 by measuring the protection of cell deaths induced by oxidative stress
(OS) by
these compounds. The results indicate that NPD1 provided the maximum
protection (60%),
followed by elovanoids at intermediate level (55%), and VLC-PUFA (50%), the
least
compared to OS (90%). (B) Elovanoid precursors protect human retinal pigment
epithelial
cells deficient in 15-LOX-1, unlike DHA, from oxidative stress conditions.
This experiment
clearly shows that VLC-PUFA, elovanoid precursors 32:6 and 34:6, and NPD1
protect
against cell death in 15-LOX-D1 cells under oxidative stress conditions. On
the other hand,
DHA was unable to do so, as the 15-LOX-D1 cells lack the enzyme required for
conversion
of DHA to the neuroprotective agent.
Figure 5. (A) Effect of PD146176 on VLC-PUFA-inhibited apoptosis induced by OS
in ARPE-19 cells. This experiment demonstrated the effect of 15-lipooxygenese
inhibitor
PD146176 on the VLC-PUFA -mediated inhibition of cell death in ARPE-19 cells
under
stressed condition. It is evident that 32:6 and 34:6 LCAF were able to induce
a substantial
amount (55 and 48% respectively) of neuroprotection compared to PD 146176
(22%) when
the stressed cells were treated with 5pm of 15-LOX-D1 inhibitor. It can be
concluded that
since PD146176 is the inhibitor of 15-lipooxygenase enzyme, it might be
protecting the
stressed RPE cells by accumulating neuroprotective agents inside the cells.
(B) Comparison
of cytoprotective capacities of NPD1, C32:6 and C34:6 VLC-PUFA on oxidative
stress-
induced apoptosis in 15-LOX-1 cells. Shown here is the comparison of
neuroprotection in a
15-LOX-1-deficient cell line under oxidative stress with 32:6 and 34:6 VLC-
PUFA along with
NPD1. 32:6 and 34:6 LCAF were able to induce neuroprotection (45% and 40%
respectively), as compared to oxidative stress (90%) under this condition. (C)
Concentration
dependent cytoprotection by C32:6 and C34:6 VLC-PUFA in oxidative-stress
induced ARPE-
19 cells. A concentration (50-500nM) kinetic of cytoprotection induced by LCAF
32:6 and
34:6 in RPE cell culture under OS was shown here. The result indicates that
there was a
gradual decrease of cell deaths starting from 50nM concentrations of both 32:6
and 34:6
LCAF, very good intermediate effect at 250nM, and maximum effect at 500nM. We
decided
to use 250nM concentrations of 32:6 and 34:6 LCAF in subsequent experiments.
(D)
Selected images of alive and dead cells from this study (control, OS,
treatment with C32:6).
Figure 6. (A) VLC-PUFA and elovanoids ELV1 and ELV2 mediated effect on Bid
upregulation in ARPE-19 cells under stress. This figure displays the
downregulation of the
proapoptotic protein of the BcI2 family Bid by western blot analysis by VLC-
PUFA and
elovanoids in RPE cells in culture under oxidative-stress. Results indicate
that upregulated
5

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Bid protein by OS, as evident from the figure, was inhibited by both
elovanoids and VLC-
PUFA. It is interesting to see that the sodium salts of the elovaniod
precursors are more
effective than the methyl ester forms. (B) VLC-PUFA and ELV1 and ELV2
compounds
mediated upregulation of Bid in ARPE-19 cells under stress. This Figure shows
the
quantification of Bid downregulation.
Figure 7. (A) VLC-PUFA and ELV1 and ELV2 compounds mediated downregulation
of Bim in ARPE-19 cells under stress. Bim, another class of BcI2 family, has
been tested like
Bid in this figure to confirm our previous results. VLC-PUFA and elovanoids
protected
against the upregulation of Bim by OS, similar to Bid, in RPE cells under
stress. (B) VLC-
PUFA and elovanoids mediated effect on Bim downregulation in ARPE-19 cells
under
stress. This Figure shows the quantification of Bim downregulation.
Figure 8. (A) Bc1-xL-upregulation by elovanoids ELV1 and ELV2 in ARPE-19 cells

under stress. BcI-xL is the antiapoptotic BcI2 family protein. Like
proapoptotic proteins Bid
and Bim, the effect of elovaniod precursors on the antiapoptotic protein BcI-
xL was tested in
this figure in RPE cells under OS. Results showed that elovaniod precursors
were able to
upregulate the BcI-xL protein in RPE cells under stress, which is the opposite
effect of Bid
and Bim. (B) Effect of NPD1, ELV1 and ELV2 on Bax expression in LOX-D cells
under
stress. Proapoptotic Box was tested in this figure. It is evident that
elovaniod precursors
downregulated the Box upregulation by OS in RPE cells under OS, which is
consistent with
our inhibition of apoptosis experiments, as shown before. C) VLC-PUFA and
elovanoids
ELV1 and ELV2 mediated effect on Box upregulation in ARPE-19 cells under
stress. In this
experiment, elovanoid precursors along with VLC-PUFA were tested on the
downregulation
of the Bax protein in RPE cells under stress.
Figure 9. (A) VLC-PUFA and elovanoids ELV1 and ELV2 mediated effect on BcI2
upregulation in ARPE-19 cells under stress. In this experiment we tested the
effect of
elovanoid precursors on BcI2 upregulation along with VLC-PUFA in stressed RPE.
(B)
Quantification of BcI2 upregulation by NPD1, ELV1 and ELV2 in LOX-D cells.
BcI2 is an
important antiapoptotic protein of the BcI2 family protein. It is evident that
elovaniod
precursors upregulated the BcI2 protein in RPE cells under stress.
Figure 10. (A) Effect of NPD1 and VLC-PUFA C32:6 and C34:6 in mediating
upregulation of SIRT1 in ARPE-19 cells. (B) Quantification of SI RT1
upregulation by NPD1,
C32:6 and C34:6. SIRT1 (Sirtuin1) belongs to a family of highly conserved
proteins linked to
caloric restriction beneficial outcomes and aging by regulating energy
metabolism, genomic
stability and stress resistance. SIRT1 is a potential therapeutic target in
several diseases
.. including cancer, diabetes, inflammatory disorders, and neurodegenerative
diseases or
disorders. Elovanoids induce cell survival involving the upregulation of the
BcI2 class of
survival proteins and the downregulation of pro-apoptotic Bad and Bax under
oxidative
6

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
stress (OS) in RPE cells. The data in this Figure suggest that elovanoids
upregulate SI RT1
abundance in human RPE cells when confronted with OS. As a consequence,
remarkable
cell survival takes place. This target of elovanoids might be relevant to
counteract
consequences of several diseases associated with SIRT1.
Figure 11. The elovanoid ELV2 in 200 nM concentrations protects neuronal cells
in
primary cultures from NMDA-induced toxicity (A), and MK-801 potentiates
protection as
assessed by MTT assay for cell viability (B). In several neurological and
neurodegenerative
diseases, such as stroke, epilepsy, status epilepticus, traumatic head injury,
etc., as well as
ophthalmological diseases, such as glaucoma, an excessive presynaptic release
of the
.. excitatory neurotransmitter glutamate takes place. As a consequence,
glutamate
transporters that function to remove extracellular glutamate from astrocytes
and neurons are
overwhelmed and the NMDA-type glutamate receptor is over-activated. This
receptor is a
calcium channel that therefore leads to a flooding of calcium into the
postsynaptic cell. The
overall phenomena is refer to as excitotoxicity that in turn leads to neuronal
damage and cell
death. MK801 is a known blocker of this receptor used here as a control. The
results in this
Figure demonstrate that when NMDA in increasing concentrations is added to
neuronal
cultures it leads to cell death, while the use of ELV2 reduces cell death and
increases cell
viability. These data support the use of the elovanoids for the treatment of
neurodegenerative diseases and conditions involving NMDA-related
excitotoxicity, such as:
.. ischemic stroke, Alzheimer's disease, Parkinson's disease, etc.
Figure 12. Elovanoids ELV2-Na and ELV2-Me are more active than DHA and NPD1
in a model of ischemic stroke after 2hrs of MCAo (middle cerebral occlusion).
To test the
novel elovanoids the experimental design consisted injecting the compounds
into the right
cerebral ventricle (5 pg/per rat, ICV), one hour after two hours of an
ischemic stroke in rats
and following thereafter the neurological behavior (neurological score) during
7 days. The
protocol in brief was as follows. The injection was made through a surgically
implanted metal
canula (Alzet) into the right lateral ventricle. Two days later the right
middle cerebral artery
(MCA) was occluded for 2 h by means of an intraluminal nylon filament (Belayev
et al,
Traslational Stroke Research, 2010). Then one hour after the compounds were
injected
dissolved in sterile cerebrospinal fluid. The occlusion was transient
performed as follows.
The right common carotid artery was exposed through an incision in the neck
and was
isolated from surrounding tissues. The distal external carotid artery and
pterygopalatine
arteries were tied. A 4-cm of 3-0 monofilament nylon suture coated with poly-
Lysine was
introduced into the internal carotid artery and MCA. The suture position was
confirmed by
advancing the suture 20-22 mm from the common carotid artery bifurcation.
Then, the rats
were allowed to awaken from anesthesia and returned to their cages. The degree
of stroke
injury was assessed by neurological assessment of each rat at 60 min after
onset of MCAo.
7

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Rats that do not demonstrate high-grade contralateral deficit (score, 10-11)
were excluded
from the study. After 2 hours of MCA occlusion, the rats were re-anesthetized
temperature
probes were reinserted and the intraluminal suture was removed. The neck
incision was
closed with silk sutures, and the animals were allowed free access to food and
water. These
results show that the use elovanoids after the ischemic event result in
remarkable
neuroprotection, suggesting a potential therapeutic benefit for the treatment
of ischemic
stroke and other neurodegenerative diseases or disorders. The sodium salt ELV2-
Na
showed a greater potency, presumably because it delivers the active form of
ELV2 and has
a more immediate effect. The methyl ester ELV2-Me is expected to first be
hydrolyzed via
.. the actions of esterases, and it may have a more delayed effect, which may
be beneficial for
a sustainable long-term treatment. Overall, these data demonstrate the
neuroprotective
effects of the elovanoids, either as pharmaceutically acceptable salts (e.g.
ELV2-Na), or in
the form of a prodrug, such as an ester derivative (e.g. ELV2-Me), or as a
pharmaceutical
composition containing a combination of the two forms that can have both an
immediate and
a sustainable long-term therapeutic effect.
Figure 13. Role of omega-3 very long chain-polyunsaturated fatty acids (n3-VLC-

PUFAs) results in the survival of photoreceptor cells, the protection of the
retina, and the
prevention of sight loss (Nat. Commun. 2015;6:1-14). Daily photoreceptor outer
segment
shed their tips that in turn are taken ¨up and digested in the phago-lysosomal
system of the
retinal pigment epithelial cell. Omega -3 fatty acids from outer segment
membrane
phospholipids are shuttled back to the inner segment of the photoreceptor to
be incorporated
again into phospholipids for membrane biogenesis of the outer segment. So
there is
recycling of this essential fatty acid and conservation during a process
called photoreceptor
outer segment renewal. We postulate that the VLC-PUFAs in the RPE cells
directly or upon
enzymatic conversion into hydroxylated derivatives, foster integrity of the
cell and as result of
the photoreceptors. This diagram depicts the desaturation and elongation steps
in the
generation of VLC-PUFAs as these molecules traffic through the endoplasmic
reticulum and
the peroxisome of the hepatocyte, the endoplasmic reticulum of the
photoreceptor inner
segment and into the photoreceptor outer segment. The elongation steps
catalyzed by
.. ELOVL4 are highlighted in red. RPE retrieval of DHA (C22:6) and of the VLC-
PUFAs from
shed photoreceptor apical disk membranes is followed by recycling of DHA and
of the VLC-
PUFAs back to the photoreceptor inner segment.
DETAILED DESCRIPTION
Before the present disclosure is described in greater detail, it is to be
understood that
this disclosure is not limited to particular embodiments described, and as
such may, of
course, vary. It is also to be understood that the terminology used herein is
for the purpose
8

of describing particular embodiments only, and is not intended to be limiting,
since the scope of
the present disclosure will be limited only by the appended claims.
Where a range of values is provided, it is understood that each intervening
value, to the
tenth of the unit of the lower limit unless the context clearly dictates
otherwise, between the
.. upper and lower limit of that range and any other stated or intervening
value in that stated range,
is encompassed within the disclosure. The upper and lower limits of these
smaller ranges may
independently be included in the smaller ranges and are also encompassed
within the
disclosure, subject to any specifically excluded limit in the stated range.
Where the stated
range includes one or both of the limits, ranges excluding either or both of
those included limits
.. are also included in the disclosure.
Unless defined otherwise, all technical and scientific terms used herein have
the same
meaning as commonly understood by one of ordinary skill in the art to which
this disclosure
belongs. Although any methods and materials similar or equivalent to those
described herein
can also be used in the practice or testing of the present disclosure, the
preferred methods and
materials are now described.
The citation of any publication is for its disclosure prior to the filing date
and should not
be construed as an admission that the present disclosure is not entitled to
antedate such
publication by virtue of prior disclosure. Further, the dates of publication
provided could be
different from the actual publication dates that may need to be independently
confirmed.
As will be apparent to those of skill in the art upon reading this disclosure,
each of the
individual embodiments described and illustrated herein has discrete
components and features
which may be readily separated from or combined with the features of any of
the other several
embodiments without departing from the scope or spirit of the present
disclosure. Any recited
method can be carried out in the order of events recited or in any other order
that is logically
.. possible.
Embodiments of the present disclosure will employ, unless otherwise indicated,

techniques of medicine, organic chemistry, biochemistry, molecular biology,
pharmacology,
toxicology, and the like, which are within the skill of the art. Such
techniques are explained fully
in the literature.
It must be noted that, as used in the specification and the appended claims,
the singular
forms "a," "an," and "the" include plural referents unless the context clearly
dictates otherwise.
Thus, for example, reference to "a support" includes a plurality of supports.
In
9
Date Regue/Date Received 2022-07-22

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
this specification and in the claims that follow, reference will be made to a
number of terms
that shall be defined to have the following meanings unless a contrary
intention is apparent.
As used herein, the following terms have the meanings ascribed to them unless
specified otherwise. In this disclosure, "comprises," "comprising,"
"containing" and "having"
and the like can have the meaning ascribed to them in U.S. patent law and can
mean"
includes," "including," and the like; "consisting essentially of" or "consists
essentially" or the
like, when applied to methods and compositions encompassed by the present
disclosure
refers to compositions like those disclosed herein, but which may contain
additional
structural groups, composition components or method steps (or analogs or
derivatives
thereof as discussed above). Such additional structural groups, composition
components or
method steps, etc., however, do not materially affect the basic and novel
characteristic(s) of
the compositions or methods, compared to those of the corresponding
compositions or
methods disclosed herein.
Prior to describing the various embodiments, the following definitions are
provided
and should be used unless otherwise indicated.
Definitions
As used herein, the nomenclature alkyl, alkoxy, carbonyl, etc. is used as is
generally
understood by those of skill in the chemical art. As used in this
specification, alkyl groups
can include straight-chained, branched and cyclic alkyl radicals containing up
to about 20
carbons, or 1 to 16 carbons, and are straight or branched. Exemplary alkyl
groups herein
include, but are not limited to, methyl, ethyl, propyl, isopropyl, isobutyl, n-
butyl, sec-butyl,
tert-butyl, isopentyl, neopentyl, tert-pentyl and isohexyl. As used herein,
lower alkyl refer to
carbon chains having from about 1 or about 2 carbons up to about 6 carbons.
Suitable alkyl
groups may be saturated or unsaturated. Further, an alkyl may also be
substituted one or
more times on one or more carbons with substituents selected from a group
consisting of
C1-C15 alkyl, ally!, allenyl, alkenyl, C3-07 heterocycle, aryl, halo, hydroxy,
amino, cyano,
oxo, thio, alkoxy, formyl, carboxy, carboxamido, phosphoryl, phosphonate,
phosphonamido,
sulfonyl, alkylsulfonate, arylsulfonate, and sulfonamide. Additionally, an
alkyl group may
contain up to 10 heteroatoms, in certain embodiments, 1, 2, 3, 4, 5, 6, 7, 8
or 9 heteroatom
substituents. Suitable heteroatoms include nitrogen, oxygen, sulfur and
phosphorous.
As used herein, "cycloalkyl" refers to a mono- or multicyclic ring system, in
certain
embodiments of 3 to 10 carbon atoms, in other embodiments of 3 to 6 carbon
atoms. The
ring systems of the cycloalkyl group may be composed of one ring or two or
more rings
which may be joined together in a fused, bridged or spiro-connected fashion.
As used herein, "aryl" refers to aromatic monocyclic or multicyclic groups
containing
from 3 to 16 carbon atoms. As used in this specification, aryl groups are aryl
radicals, which
may contain up to 10 heteroatoms, in certain embodiments, 1, 2, 3 or 4
heteroatoms. An

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
aryl group may also be optionally substituted one or more times, in certain
embodiments, 1
to 3 or 4 times with an aryl group or a lower alkyl group and it may be also
fused to other aryl
or cycloalkyl rings. Suitable aryl groups include, for example, phenyl,
naphthyl, tolyl,
imidazolyl, pyridyl, pyrroyl, thienyl, pyrimidyl, thiazolyl and furyl groups.
As used in this specification, a ring is defined as having up to 20 atoms that
may
include one or more nitrogen, oxygen, sulfur or phosphorous atoms, provided
that the ring
can have one or more substituents selected from the group consisting of
hydrogen, alkyl,
allyl, alkenyl, alkynyl, aryl, heteroaryl, chloro, iodo, bromo, fluoro,
hydroxy, alkoxy, aryloxy,
carboxy, amino, alkylamino, dialkylamino, acylamino, carboxamido, cyano, oxo,
thio,
alkylthio, arylthio, acylthio, alkylsulfonate, arylsulfonate, phosphoryl,
phosphonate,
phosphonamido, and sulfonyl, and further provided that the ring may also
contain one or
more fused rings, including carbocyclic, heterocyclic, aryl or heteroaryl
rings.
As used herein, alkenyl and alkynyl carbon chains, if not specified, contain
from 2 to
carbons, or 2 to 16 carbons, and are straight or branched. Alkenyl carbon
chains of from
15 .. 2 to 20 carbons, in certain embodiments, contain 1 to 8 double bonds,
and the alkenyl
carbon chains of 2 to 16 carbons, in certain embodiments, contain Ito 5 double
bonds.
Alkynyl carbon chains of from 2 to 20 carbons, in certain embodiments, contain
1 to 8 triple
bonds, and the alkynyl carbon chains of 2 to 16 carbons, in certain
embodiments, contain 1
to 5 triple bonds.
20 As used herein, "heteroaryl" refers to a monocyclic or multicyclic
aromatic ring
system, in certain embodiments, of about 5 to about 15 members where one or
more, in one
embodiment 1 to 3, of the atoms in the ring system is a heteroatom, that is,
an element other
than carbon, including but not limited to, nitrogen, oxygen or sulfur. The
heteroaryl group
may be optionally fused to a benzene ring. Heteroaryl groups include, but are
not limited to,
furyl, imidazolyl, pyrrolidinyl, pyrimidinyl, tetrazolyl, thienyl, pyridyl,
pyrrolyl, N-methylpyrrolyl,
quinolinyl and isoquinolinyl.
As used herein, "heterocycly1" refers to a monocyclic or multicyclic non-
aromatic ring
system, in one embodiment of 3 to 10 members, in another embodiment of 4 to 7
members,
in a further embodiment of 5 to 6 members, where one or more, in certain
embodiments, 1 to
3, of the atoms in the ring system is a heteroatom, that is, an element other
than carbon,
including but not limited to, nitrogen, oxygen or sulfur. In embodiments where
the
heteroatom(s) is(are) nitrogen, the nitrogen is optionally substituted with
alkyl, alkenyl,
alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl, cycloalkyl, heterocyclyl,
cycloalkylalkyl,
heterocyclylalkyl, acyl, guanidino, or the nitrogen may be quaternized to form
an ammonium
group where the substituents are selected as above.
As used herein, "aralkyl" refers to an alkyl group in which one of the
hydrogen atoms
of the alkyl is replaced by an aryl group.
11

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
As used herein, "halo", "halogen" or "halide" refers to F, Cl, Br or I.
As used herein, "haloalkyl" refers to an alkyl group in which one or more of
the
hydrogen atoms are replaced by halogen. Such groups include, but are not
limited to,
chloromethyl and trifluoromethyl.
As used herein, "aryloxy" refers to RO-, in which R is aryl, including lower
aryl, such
as phenyl.
As used herein, "acyl" refers to a -COR group, including for example
alkylcarbonyl,
cycloalkylcarbonyl, arylcarbonyl, or heteroarylcarbonyls, all of which may be
optionally
substituted.
As used herein, "0)-3", "(5-6", etc. refers to the customary nomenclature of
polyunsaturated fatty acids or their derivatives, wherein the position of a
double bond (C=C)
is at the carbon atom counted from the end of the carbon chain (methyl end) of
the fatty acid
or fatty acid derivative. For example, "o)-3" means the third carbon atom from
the end of the
carbon chain of the fatty acid or fatty acid derivative. Similarly, "@-3", "@-
6", etc. also refers
to the position of a substituent such as a hydroxyl group (OH) located at a
carbon atom of
the fatty acid or fatty acid derivative, wherein the number (e.g. 3, 6, etc.)
is counted from the
end of the carbon chain of the fatty acid or fatty acid derivative.
As used herein, the abbreviations for any protective groups and other
compounds,
are, unless indicated otherwise, in accord with their common usage, recognized
abbreviations, or the IUPAC-IUB Commission on Biochemical Nomenclature (see,
(1972)
Biochem. 11:942-944).
As used herein, wherein in chemical structures of the compounds of the
disclosure
are shown having a terminal carboxyl group "-COOR" the "R" is intended to
designate a
group covalently bonded to the carboxyl such as an alkyl group. In the
alternative, the
carboxyl group is further intended to have a negative charge as "-COO- and R
is a cation
including a metal cation, an ammonium cation and the like.
As used herein "subject" is an animal, typically a mammal, including human,
such as
a patient.
As used herein, "pharmaceutically acceptable derivatives" of a compound
include
salts, esters, enol ethers, enol esters, acetals, ketals, orthoesters,
hemiacetals, hemiketals,
acids, bases, solvates, hydrates or prodrugs thereof. Such derivatives may be
readily
prepared by those of skill in this art using known methods for such
derivatization. The
compounds produced may be administered to animals or humans without
substantial toxic
effects and either are pharmaceutically active or are prodrugs.
Pharmaceutically acceptable
salts include, but are not limited to, amine salts, such as but not limited to
N,N'-
dibenzylethylenediamine, chloroprocaine, choline, ammonia, diethanolamine and
other
12

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
hydroxyalkylamines, ethylenediamine, N-methylglucamine, procaine, N-
benzylphenethylamine, 1-para-chlorobenzy1-2-pyrrolidin-1'-
ylmethylbenzimidazole,
diethylamineand other alkylamines, piperazine and
tris(hydroxymethyl)aminomethane; alkali
metal salts, such as but not limited to lithium, potassium and sodium; alkali
earth metal salts,
such as but not limited to barium, calcium and magnesium; transition metal
salts, such as
but not limited to zinc; and other metal salts, such as but not limited to
sodium hydrogen
phosphate and disodium phosphate; and also including, but not limited to,
salts of mineral
acids, such as but not limited to hydrochlorides and sulfates; and salts of
organic acids, such
as but not limited to acetates, lactates, malates, tartrates, citrates,
ascorbates, succinates,
butyrates, valerates and fumarates. Pharmaceutically acceptable esters
include, but are not
limited to, alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl, heteroaralkyl,
cycloalkyl and
heterocyclyl esters of acidic groups, including, but not limited to,
carboxylic acids, phosphoric
acids, phosphinic acids, sulfonic acids, sulfinic acids and boronic acids.
Pharmaceutically
acceptable enol ethers include, but are not limited to, derivatives of formula
C=C(OR) where
R is hydrogen, alkyl, alkenyl, alkynyl, aryl, heteroaryl, aralkyl,
heteroaralkyl, cycloalkyl ar
heterocyclyl. Pharmaceutically acceptable enol esters include, but are not
limited to,
derivatives of formula C=C(OC(0)R) where R is hydrogen, alkyl, alkenyl,
alkynyl, aryl,
heteroaryl, aralkyl, heteroaralkyl, cycloalkyl ar heterocyclyl.
Pharmaceutically acceptable
solvates and hydrates are complexes of a compound with one or more solvent or
water
molecules, or 1 to about 100, or 1 to about 10, or one to about 2, 3 or 4,
solvent or water
molecules.
As used herein, amelioration of the symptoms of a particular disorder by
administration of a particular compound or pharmaceutical composition refers
to any
lessening, whether permanent or temporary, lasting or transient that can be
attributed to or
associated with administration of the composition.
The term "therapeutically effective amount" as used herein refers to that
amount of
an embodiment of the composition or pharmaceutical composition being
administered that
will relieve to some extent one or more of the symptoms of the disease or
condition being
treated, and/or that amount that will prevent, to some extent, one or more of
the symptoms
of the condition or disease that the subject being treated has or is at risk
of developing. As
used interchangeably herein, "subject," "individual," or "patient," refers to
a vertebrate,
preferably a mammal, more preferably a human. Mammals include, but are not
limited to,
murines, simians, humans, farm animals, sport animals, and pets. The term
"pet" includes a
dog, cat, guinea pig, mouse, rat, rabbit, ferret, and the like. The term farm
animal includes a
horse, sheep, goat, chicken, pig, cow, donkey, llama, alpaca, turkey, and the
like.
A "pharmaceutically acceptable excipient," "pharmaceutically acceptable
diluent,"
"pharmaceutically acceptable carrier," or "pharmaceutically acceptable
adjuvant" means an
13

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
excipient, diluent, carrier, and/or adjuvant that are useful in preparing a
pharmaceutical
composition that are generally safe, non-toxic and neither biologically nor
otherwise
undesirable, and include an excipient, diluent, carrier, and adjuvant that are
acceptable for
veterinary use and/or human pharmaceutical use. "A pharmaceutically acceptable
excipient,
diluent, carrier and/or adjuvant" as used in the specification and claims
includes one and
more such excipients, diluents, carriers, and adjuvants.
As used herein, a "pharmaceutical composition" or a "pharmaceutical
formulation" is
meant to encompass a composition or pharmaceutical composition suitable for
administration to a subject, such as a mammal, especially a human and that
refers to the
combination of an active agent(s), or ingredient with a pharmaceutically
acceptable carrier or
excipient, making the composition suitable for diagnostic, therapeutic, or
preventive use in
vitro, in vivo, or ex vivo. In general a "pharmaceutical composition" is
sterile, and preferably
free of contaminants that are capable of eliciting an undesirable response
within the subject
(e.g., the compound(s) in the pharmaceutical composition is pharmaceutical
grade).
.. Pharmaceutical compositions can be designed for administration to subjects
or patients in
need thereof via a number of different routes of administration including
oral, intravenous,
buccal, rectal, parenteral, intraperitoneal, intradermal, intracheal,
intramuscular,
subcutaneous, inhalational and the like.
The term "administration" refers to introducing a composition of the present
disclosure into a subject. One preferred route of administration of the
composition is topical
administration. However, any route of administration, such as oral,
intravenous,
subcutaneous, peritoneal, intra-arterial, inhalation, vaginal, rectal, nasal,
introduction into the
cerebrospinal fluid, or instillation into body compartments can be used.
As used herein, "treatment" and "treating" refer to the management and care of
a
subject for the purpose of combating a condition, disease or disorder, in any
manner in
which one or more of the symptoms of a disease or disorder are ameliorated or
otherwise
beneficially altered. The term is intended to include the full spectrum of
treatments for a
given condition from which the patient is suffering, such as administration of
the active
compound for the purpose of: alleviating or relieving symptoms or
complications; delaying
the progression of the condition, disease or disorder; curing or eliminating
the condition,
disease or disorder; and/or preventing the condition, disease or disorder,
wherein
"preventing" or "prevention" is to be understood to refer to the management
and care of a
patient for the purpose of hindering the development of the condition, disease
or disorder,
and includes the administration of the active compounds to prevent or reduce
the risk of the
onset of symptoms or complications. The patient to be treated is preferably a
mammal, in
particular a human being. Treatment also encompasses any pharmaceutical use of
the
compositions herein, such as use for treating a disease as provided herein.
14

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Discussion
Inflammatory, degenerative, and neurodegenerative diseases include a large
number
of diseases that affect a very large number of people worldwide. In most
cases, these
diseases and related conditions and disorders are difficult to treat, and
remain as an unmet
medical need.
Inflammatory diseases in the scope of this disclosure include acute and
chronic
disorders where homeostasis is disrupted by an abnormal or dysregulated
inflammatory
response. These conditions are initiated and mediated by a number of
inflammatory factors,
including oxidative stress, chemokines, cytokines, breakage of blood/tissue
barriers,
autoimmune diseases or other conditions that engage leukocytes,
monocytes/macrophages
or parenchymal cells that induce excessive amounts of pro-cell injury, pro-
inflammatory/disruptors of homeostasis mediators. These diseases occur in a
wide range of
tissues and organs and are currently treated, by anti-inflammatory agents such
as
corticosteroids, non-steroidal anti-inflammatory drugs, TNF modulators, COX-2
inhibitors,
etc.
Degenerative diseases include conditions that involve progressive loss of
vital cells
and tissues that result in progressive impairment of function, such as loss of
cartilage in
knees, hip joints or other joints such as in osteoarthritis. Other
degenerative diseases
engages cellular and intercellular homeostasis perturbations and includes
heart disease,
atherosclerosis, cancer, diabetes, intestinal bowel disease, osteoporosis,
prostatitis,
rheumatoid arthritis, etc.
Neurodegenerative diseases include some of the major diseases of the brain,
retina,
spinal cord and peripheral nerves, whereby a progressive demise of cellular
organization
leads to impaired function. These are due to immune or inflammatory disorders
and/or to
inherited conditions or aging. They include multiple sclerosis, Alzheimer's
disease,
Parkinson's disease, amyotrophic lateral sclerosis, retina degenerative
diseases such as
age-related macular degeneration, inherited eye diseases such as retinitis
pigmentosa,
glaucoma, etc.
Retinal degenerative diseases are the leading causes of blindness that affects
very
large numbers of people. Retinal degeneration is the deterioration of the
retina caused by
the progressive and eventual death of the photoreceptor cells of the retina.
Examples of
common retinal degenerative diseases include retinitis pigmentosa, age-related
macular
degeneration, and Stargardt disease. Retinitis pigmentosa affects between
50,000 and
100,000 people in the United States alone, and macular degeneration is the
leading cause of
vision loss for those aged 55 and older in the United States, affecting more
than 10 million
people. There are no effective treatments for these and other retinal
degenerative diseases.

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Despite progress made in understanding the pathophysiology of inflammatory and

degenerative diseases, the detailed molecular mechanisms involved in the
initiation and
progression of these conditions remain poorly understood. For retinal
degenerative
diseases, the detailed molecular mechanisms involved in the progressive loss
of
photoreceptor cells remain unknown, and available treatments today are not
able to
effectively treat these major diseases and prevent loss of sight. What is
needed is a method
for the prevention and treatment of retinal degenerative diseases that ensures
the survival of
the retina photoreceptor cells.
Available treatments today are not able to effectively treat these major
diseases or to
slow-down their progressive impairment of vital functions. What is needed is a
method that
ensures the survival of critical cells undergoing oxidative stress or other
homeostatic
disruptions. Therefore, there is a major therapeutic void for the management
of
inflammatory, neuroinflammatory, degenerative and neurodegenerative diseases.
This disclosure provides compounds, compositions and methods for the effective
prevention and treatment of inflammatory and degenerative diseases, including
neurodegenerative diseases and retinal degenerative diseases. The disclosure
is based on
new findings regarding the key protective role of certain omega-3 very long
chain-
polyunsaturated fatty acids (n3 VLC-PUFA) and related hydroxylated
derivatives.
In particular, described herein are methods and compounds for the protection
of the
retina by inducing the survival of photoreceptors. The methods describe herein
involve the
use of compounds that induce survival signaling in both the retinal pigment
epithelial cells
and photoreceptors.
Recent investigations have shown that certain polyunsaturated fatty acids
(PUFA)
are enzymatically converted to bioactive derivatives that play important roles
in inflammation
and related conditions. Notable among these are the omega-3 (n3) fatty acids
containing 22
carbons including eicosapentaenoic acid (EPA or C20:5n3) (20 carbons, 5 double
bonds,
omega-3), docosapentaenoic acid (DPA or 022:5n3), and especially
docosahexaenoic acid
(DHA or C22:6n3) (22 carbons, 6 double bonds, omega-3). These PUFA are
converted via
lipoxygenase-type enzymes to biologically active hydroxylated PUFA
derivatives. Most
important among these are specific types of hydroxylated derivatives that are
generated in
certain inflammation-related cells via the action of a lipoxygenase (LO)
enzyme (e.g. 15-LO,
12-L0), and result in the formation of mono-, di- or tri-hydroxylated PUFA
derivatives with
potent actions including anti-inflammatory, pro-resolving, neuroprotective or
tissue-protective
actions, among others. For example, neuroprotectin D1 (NPD1), a dihydroxy
derivative from
DHA formed in cells via the enzymatic action of 15-lipoxygenase (15-LO) was
shown to have
a defined R/S and Z/E stereochemical structure (10R,17S-dihydroxy-docosa-
4Z,7Z,11E,
13E,15Z,19Z-hexaenoic acid) and a unique biological profile that includes
stereoselective
16

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
potent anti-inflammatory, homeostasis-restoring, pro-resolving, bioactivity.
NPD1 has been
shown to modulate neuroinflammatory signaling and proteostasis, and to promote
nerve
regeneration, neuroprotection, and cell survival.
Other important types of omega-3 fatty acids are the omega-3 very-long-chain
polyunsaturated fatty acids (n3 VLC-PUFA or VLC-PUFA), which are produced in
cells
containing elongase enzymes that elongate PUFA with lower number of carbons to
VLC-
PUFA containing between 24 to 36 carbons. Representative types of VLC-PUFA
include
C32:6n3 (32 carbons, 6 double bonds, omega-3), C34:6n3, C32:5n3, and 034:5n3,
which
are biogenically derived through the action of elongase enzymes, particularly
ELOVL4
(ELOngation of Very Long chain fatty acids 4). These fatty acids are also
acylated in
complex lipids including sphingolipids and phospholipids particularly in
certain molecular
species of phosphatidyl choline. These VLC-PUFA are thought to display
functions in
membrane organization, and their significance to health is increasingly
recognized. The
biosynthesis and biological functions of VLC-PUFA have been the subject of a
number of
recent investigations that have suggested potential roles in certain diseases.
An increasing number of studies have demonstrated the importance of VLC-PUFA
in
the retina, an integral part of the central nervous system. For example, the
autosomal
dominant Stargardt-like macular dystrophy (STGD3), a Juvenile-onset retinal
degenerative
disease is caused by mutations in exon 6 of the ELOVL4 gene that leads to a
truncated
ELOVL4 protein (a key elongase enzyme) without an endoplasmic reticulum (ER)
retention/retrieval signal, resulting in severe decrease in the biosynthesis
of VLC-PUFA. Low
retinal levels of VLC-PUFA and abnormally low n3/n6 ratios also occur in age-
related
macular degeneration (AMD) donor eyes as compared to age-matched control eye
donors.
Recessive ELOVL4 mutations display clinical features of ichthyosis, seizures,
mental
retardation, and spastic quadriplegia that resembles Sjogren-Larsson syndrome
(SLS) with
severe neurologic phenotype implying the significance of VLC-PUFA synthesis
for the
central nervous system and cutaneous development.
VLC-PUFA were found to be incorporated in phospholipids of the photoreceptor
outer
membrane, and were shown to play important roles in the longevity of
photoreceptors, and in
their synaptic function and neuronal connectivity. Therefore, bioactive
derivatives based on
VLC-PUFA, which are able to prevent the apoptosis of photoreceptor cells may
provide
therapeutic benefits for various types of retinal degenerative diseases,
including Stargardt-
like macular dystrophy (STGD3), and X-linked juvenile retinoschisis (XLRS) an
inherited
early onset retinal degenerative disease caused by mutations in the RS1 gene,
which is the
leading cause of juvenile macular degeneration in males. This condition
denotes a significant
photoreceptor synaptic impairment for which there is no available treatment
17

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Although VLC-PUFA are attracting increasing attention, their detailed
biological role
and functional significance remains poorly understood, and their potential use
in medicine
has not been fully appreciated. In particular, the detailed role and potential
beneficial use of
VLC-PUFA and their synthetic derivatives as potential therapeutics remains to
be
established. Moreover, the potential use of VLC-PUFA in inflammatory,
degenerative
diseases, and neurodegenerative diseases of the retina and the brain, such as
stroke,
Alzheimer's disease, autism spectrum disorders, schizophrenia, Parkinson's
disease,
remains to be developed.
The structures, properties, and potential effects of VLC-PUFA in cells and
tissues,
such as the retina, where they are known to play dominant roles were
evaluated.
Experiments were done using human retinal pigment epithelial (RPE) cells,
which are
neuroectoderm-derived post-mitotic cells of the retina, an integral part of
the central nervous
system. These cells are richly endowed with a multitude of mechanisms to
protect
themselves from injury and to protect other cells, particularly the survival
of photoreceptors.
They are the most active phagocyte of the human body, critical for the health
of
photoreceptors and vision, and have the ability to secrete neurotrophins and
other beneficial
substances. In pathological conditions they recapitulate aspects of
Alzheimer's disease by
processing amyloid precursor protein and contributing to the formation of
Drusen,
analogously to the senile amyloid plaques. Thus, these are among the reasons
that some of
the experimental data included in this disclosure were obtained with RPE
cells. Therefore,
the data provided herein are representative of the expected activities of the
provided
compounds in other cells and tissues where VLC-PUFA are known to be generated
or be
present. Based on the data detailed herein, we postulate that VLC-PUFA are
expressed in
certain forms of these cells, and in a paracrine fashion they induce the
expression of
protective phonotypes of these cells. These cells appear between the RPE and
the
photoreceptors, a zone of immune privilege regulated by immunosuppressive RPE
signals
and other factors.
There is a growing evidence that a reduced presence of VLC-PUFA in certain
cells
and tissues is associated with degenerative, neurodegenerative, and retinal
degenerative
diseases, which are linked to excessive and persistent inflammatory
environment.
The naturally occurring VLC-PUFA are biosynthesized via the actions of
elongase
enzymes, such as ELOVL4, which add two carbons at a time starting from DHA
(which has
22 carbon atoms), as summarized in Figure 13. Thus, biogenetically derived VLC-
PUFA
contain only an even number of carbons ranging from 24 of up to 42 carbons.
Such
naturally occurring VLC-PUFA have been detected in the form of free acids or
as
components of cellular lipids in mammalian tissues. Due to the requirement of
elongase
18

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
enzymes in their biosynthesis, VLC-PUFA containing an odd number of carbons
are not
known to exist in nature.
This disclosure was conceived on the hypothesis that therapeutic interventions
for
these VLC-PUFA related diseases can be developed by providing
pharmacologically
effective amounts of compounds that mimic the structures and biological
activities of locally
generated VLC-PUFA. While there are several VLC-PUFA fatty acids that have
been
identified in cells and tissues, their biological roles have been presumed to
be due to these
naturally generated fatty acids and the corresponding phospholipids.
In this disclosure we describe for the first time compounds having carbon
chains
analogous to VLC-PUFA that in addition to having 6 or 5 C=C bonds, they also
contain one
or two hydroxyl groups. Based on the hypothesis that compounds of this type
may be
responsible for the protective and neuroprotective actions of VLC-PUFA, we
sought to
identify their existence in human retinal pigment epithelial cells in culture
in the presence of a
VLC-PUFA added in its fatty acid form. As shown in Figure 2, we had obtained
evidence of
the formation of mono-hydroxy and di-hydroxy VLC-PUFA derivatives with
molecular
structures that are analogous to DHA-derived 17-hydroxy-DHA and the di-hydroxy

compound NPD1 (10R,17S-dihydroxy-docosa-4Z,7Z,11E,13E,15Z,19Z-hexaenoic acid).

Given the very small (nanogram) quantities of these hydroxylated derivatives
of VLC-PUFA,
it was not possible to identify their complete structure and stereochemistry
(R or S hydroxy
groups, Z or E double bonds). Moreover, the detected compounds were not
identified from
tissues naturally occurring in nature, but from the result of an artificial
experiment combining
a human cell and a VLC-PUFA. Therefore, the natural occurrence of the provided
mono- and
di-hydroxylated is not known at this time.
The provided compounds are not obtained from natural sources but they are
prepared by adapting methods known in the art, starting with commercially
available
materials. The provided preparation methods were designed to be suitable to
the unique
hydrophobic properties of VLC-PUFA, which differ significantly from compounds
having a
total number of carbons of 22 carbons or less.
The provided compounds are chemically modified pharmaceutically acceptable
derivatives to enhance their chemical and biological stability, and to enable
their use in
therapeutic applications involving various forms of drug delivery.
Rather than provide VLC-PUFA in the form occurring in nature, this disclosure
provides compounds that have stereochemically pure structures and are
chemically
synthesized and modified to have additional structural features and properties
that enable
them to exert pharmacological activity. The disclosure also provides
pharmacologically
effective compositions of the provided compounds that enhance their ability to
be delivered
to a subject in a manner that can reach the targeted cells and tissues.
19

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
It is currently known that:(a) Mutations in the elongase enzyme ELVOL4 leads
to
retinal degenerative diseases; (b) ELOVL4 is a key enzyme involved in the
conversion of
DHA (C22:6) into VLC-PUFA; (c) Genetic ablation of the protein that is
necessary to capture
DHA into retinal cells containing ELOVL4 products result in a drastically
decreased levels of
the VLC-PUFAs with consequent retinal degeneration; and (d) Oxidative stress
(OS) is
associated with the early stages of degenerative, neurodegenerative, and
retinal
degenerative diseases.
While not wishing to be bound by any one theory, it was considered that VLC-
PUFA
or their endogenously produced derivatives may play a direct role in neuronal
protection and
survival, which can provide the basis for a new concept for the treatment of
inflammatory,
degenerative and neurodegenerative diseases.
The present disclosure is supported by the following new and unexpected data
herein disclosed:
(a) VLC-PUFA C32:6 and C34:6 are protective against OS in RPE cells (Figures
4, 5,
6, 7, 8, 9, 10).
(b) Protection against OS by VLC-PUFA is not inhibited by inhibitors of the 15
LOX-1
enzyme (Figure 5A). Since 15 LOX-1 is associated with the conversion of DHA
into NPD1,
the observed actions of VLC-PUFA suggest that there are different enzymes
associated with
their protective role.
(c) Cell-derived hydroxylated derivatives (29-hydroxy-34:6 and 22,29-dihydroxy-
34:6)
could be detected in cultures of VLC-PUFA C34:6 from human retinal pigment
epithelial cells
in culture (Figure 2).
(d) Chemical synthesis afforded stereochemically pure di-hydroxylated
derivatives of
VLC-PUFA C32:6 and C34:6, named herein as elovanoids ELV1 and ELV2
respectively,
prepared as sodium salts or methyl esters (Figure 3).
(e) The synthetic elovanoids ELV1 and ELV2 as sodium salts or methyl esters
exhibited more potent activity against OS than the related VLC-PUFA (Figures
4).
(f) The potent activities of elovanoids ELV1 and ELV2 co-related with potent
down-
regulation of the proapoptotic proteins of the BcI2 family Bid (Figure 6), Bim
(Figure 7), Bax
(Figure 8B).
(g) The potent activities of elovanoids ELV1 and ELV2 co-related with potent
up-
regulation of the antiapoptotic proteins of the Bc12 family BcI-xL (Figure 8A)
and BcI2 (Figure
9).
(h) VLC-PUFA C32:6 and C34:6 mediate the upregulation of SIRT1 in ARPE-19
cells
(Figure 10).
(i) The elovanoid ELV2 (as the sodium salt or methyl ester) potently protects
neuronal cells in primary cultures from NMDA-induced toxicity (Figure 11).

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
(j) The synthetic elovanoids ELV2-Na and ELV2-Me were shown to have potent in
vivo neuroprotective effects in a rat model of ischemic stroke after 2 hours
of middle cerebral
occlusion (MCAo) (Figure 12). Both elovanoid derivatives exhibited greater in
vivo potency
than DHA or NPD1, suggesting a remarkable neuroprotection and a potential
therapeutic
benefit for the treatment of ischemic stroke and other neurodegenerative
diseases or
disorders.
(k) The greater potency of elovanoid ELV2 (as sodium salt or methyl ester) vs
the
docosanoids (DHA, NPD1) (Figure 12) may be due to either a different mechanism
of action,
a different metabolic profile that increases their bioavailability, or a
different localization (e.g.
intracellular receptors in the nuclear membrane) due to their longer fatty
acid length and
potentially greater hydrophobicity and structural rigidity.
(i) Taken together, the above previously unknown data, including the structure
and
activity of the elovanoids, and the potent neuroprotective activities of
elovanoid derivatives
such as ELV1 and ELV2, provide the basis for the present disclosure.
The compounds and compositions provided by this disclosure are able to restore
homeostasis and induce survival signaling in certain cells undergoing
oxidative stress or
other homeostatic disruptions. The disclosure also provides methods of use of
the provided
compounds and compositions containing a hydroxylated derivative of very long
chain
polyunsaturated fatty acids, as the free carboxylic acids or their
pharmaceutically acceptable
salts, or as their corresponding esters or other prodrug derivatives. The
provided
compounds can be readily prepared by adapting methods known in the art,
starting with
commercially available materials.
The bioactivity of the provided compounds, as exemplified by the elovanoid
derivatives ELV1 and ELV2, is attributed to their ability to reach the
targeted human cells
and exert their biological actions either by entering into the cell or/ and by
acting at a
membrane bound receptor. Alternatively, the provided compounds can act via
intracellular
receptors (e.g. nuclear membrane), and thus they would work specifically by
affecting key
signaling events.
Administration of a pharmaceutical composition, containing a provided compound
and a pharmaceutically acceptable carrier, restores the homeostatic balance
and promotes
the survival of certain cells that are essential for maintaining normal
function. The provided
compounds, compositions, and methods can be used for the preventive and
therapeutic
treatment of inflammatory, degenerative, and neurodegenerative diseases. This
disclosure
targets critical steps of the initiation and early progression of these
conditions by mimicking
the specific biology of intrinsic cellular/organs responses to attain potency,
selectivity, devoid
of side effects and sustained bioactivity.
21

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Other compositions, compounds, methods, features, and advantages of the
present
disclosure will be or become apparent to one having ordinary skill in the art
upon
examination of the following drawings, detailed description, and examples. It
is intended that
all such additional compositions, compounds, methods, features, and advantages
be
included within this description, and be within the scope of the present
disclosure.
Compounds
Described herein are compounds and compositions based on very long chain
polyunsaturated fatty acids and their hydroxylated derivatives.
In some embodiments the provided compounds and compositions are based on
compounds with the general structures of A or B, wherein n is a number
selected from a
group consisting of 0 to 19 and the compounds are carboxylic acids or their
pharmaceutically
acceptable salts. Compounds of structure A are based on very long chain
polyunsaturated
fatty acids with a total of 23 to 42 carbon atoms in the carbon chain and 6
alternating cis-
carbon-carbon double bonds starting at positions a-3 (omega-3), ro-6,
o-12, a-15 and
0)-18. Compounds of structure B are based on very long chain polyunsaturated
fatty acids
with a total of 23 to 42 carbon atoms in the carbon chain and preferably 5
alternating cis-
carbon-carbon double bonds starting at positions o)-3 (omega-3), o.)-6, co-9,
co-12 and 0)-15.
,0
OH OH
A B =*
In preferred embodiments, n is a number selected from a group consisting of 0
to 13.
In further preferred embodiments, n is a number selected from 1, 3, 5, 7, 9,
11 or 13,
and the fatty acid contains a total of 24, 26, 28, 30, 32, 34 or 36 carbon
atoms.
In other preferred embodiments, n is a number selected from a group consisting
of 0,
2, 4, 6, 8, 10 or 12, and the fatty acid contains a total of 23, 25, 27, 19,
31, 33 or 35 carbon
atoms.
In other embodiments the disclosure provides compounds that are carboxyl
derivatives of very long chain polyunsaturated fatty acids of the general
structures C or D,
wherein n is a number selected from a group consisting of 0 to 19 and the
carboxyl
derivative is an ester or a pharmaceutically acceptable salt, wherein the R
group is selected
from a group consisting of methyl, ethyl, alkyl, or a cation selected from a
group consisting
of: ammonium cation, iminium cation, or a metal cation. Compounds of structure
C are ester
derivatives of very long chain polyunsaturated fatty acids with a total of 23
to 42 carbon
22

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
atoms in the carbon chain and preferably 6 alternating cis-carbon-carbon
double bonds
starting at positions 0)-3 (omega-3), co-6, 0)-9, 0)-12, 00-15 and co-18.
Compounds of structure
D are carboxyl derivatives of very long chain polyunsaturated fatty acids with
a total of 24 to
42 carbon atoms in the carbon chain and preferably 5 alternating cis-carbon-
carbon double
bonds starting at positions co-3 (omega-3), co-6, co-9, co-12 and 0)-15.
,0 ,0
OR OR
C D
In preferred embodiments, n is a number selected from a group consisting of 0
to 13.
In further preferred embodiments, n is a number selected from 1, 3, 5, 7, 9,
11 or 13,
and the fatty acid contains a total of 24, 26, 28, 30, 32, 34 or 36 carbon
atoms.
In other preferred embodiments, n is a number selected from a group consisting
of 0,
2, 4, 6, 8, 10 or 12, and the fatty acid contains a total of 23, 25, 27, 19,
31, 33 or 35 carbon
atoms.
In some preferred embodiments, the R group is methyl or ethyl, or a metal
cation
selected from a group consisting of sodium, potassium, magnesium, zinc, or
calcium cation.
In an exemplary preferred embodiment, the present disclosure provides
compounds
of the general formula C, wherein:
n is a number selected from a group consisting of zero 1, 3, 5, 7, 9, 11 or
13, wherein
the fatty acid chain contains a total of 24, 26, 28, 30, 32, 34 or 36 carbon
atoms; and
As used herein and in other structures of the present disclosure, the
compounds of
the disclosure are shown having a terminal carboxyl group "-COOR" the "R" is
intended to
designate a group covalently bonded to the carboxyl such as an alkyl group. In
the
alternative, the carboxyl group is further intended to have a negative charge
as "-coo- and
R is a cation including a metal cation, an ammonium cation and the like.
R is selected from a group consisting of methyl, ethyl, alkyl, or a cation
selected from
a group consisting of: ammonium cation, iminium cation, or a metal cation. In
some preferred
embodiments the metal cation is selected from a group consisting of sodium,
potassium,
magnesium, zinc or calcium cation.
In some preferred embodiments, the R group is methyl or ethyl, or a metal
cation
selected from a group consisting of sodium, potassium, magnesium, zinc or
calcium cation.
In a further preferred embodiment, the present disclosure provides compounds
of the
general formula C, wherein:
23

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
n is 9 or 11, wherein the fatty acid chain contains a total of 32 or 34 carbon
atoms;
and
R is selected from a group consisting of methyl, ethyl, alkyl, or a cation
selected from
a group consisting of: ammonium cation, iminium cation, or a metal cation. In
some preferred
embodiments the metal cation is selected from a group consisting of sodium,
potassium,
magnesium, zinc, or calcium cation.
In other preferred embodiments, the carboxyl derivative is part of a glycerol-
derived
phospholipid, wherein R is a glycerol phospholipid that may contain an
additional
polyunsaturated fatty acid, as exemplified in structures E and F.
,0 ,0
(CH2)n-a<
OH
0-Nc 0
=
P
= o =
of
=
"=== "= N-r
In other embodiments the provided compounds have the general structures of G
or
H, wherein n is a number selected from a group consisting of 0 to 19, and the
carboxylate R
group is selected from a group consisting of an ester or a pharmaceutically
acceptable salt,
wherein the R group is selected from a group consisting of hydrogen, methyl,
ethyl, alkyl, or
a cation selected from a group consisting of: ammonium cation, iminium cation,
or a metal
cation. Compounds of structure G are mono-hydroxylated derivatives of very
long chain
polyunsaturated fatty acids with a total from 23 to 42 carbon atoms in the
carbon chain, a
hydroxyl group at position co-6, and with 6 carbon-carbon double bonds
starting at positions
co-3, o)-7, co-9, ro-12, 0)-15 and co-18. Compounds of structure H are mono-
hydroxylated
derivatives of very long chain polyunsaturated fatty acids with a total from
23 to 42 carbon
atoms in the carbon chain, a hydroxyl group at position (0-6, and with 5
carbon-carbon
double bonds starting at positions 0o-3, co-7, co-9, (0-12, and (0-15.
,0
(CH2)n-4(
OR OR
1
.===
G HO H HO
In preferred embodiments, n is a number selected from a group consisting of 1
to 13.
In further preferred embodiments, n is a number selected from 1, 3, 5, 7, 9,
11 or 13,
and the fatty acid contains a total of 24, 26, 28, 30, 32, 34 or 36 carbon
atoms.
24

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
In other preferred embodiments, n is a number selected from a group consisting
of 0,
2, 4, 6, 8, 10 or 12, and the fatty acid contains a total of 23, 25, 27, 19,
31, 33 or 35 carbon
atoms.
As used herein and in other structures of the present disclosure, the
compounds of
the disclosure are shown having a terminal carboxyl group "-COOR" the "R" is
intended to
designate a group covalently bonded to the carboxyl such as an alkyl group. In
the
alternative, the carboxyl group is further intended to have a negative charge
as "-coo- and
R is a cation including a metal cation, an ammonium cation and the like.
R is selected from a group consisting of methyl, ethyl, alkyl, or a cation
selected from
a group consisting of: ammonium cation, iminium cation, or a metal cation. In
some preferred
embodiments the metal cation is selected from a group consisting of sodium,
potassium,
magnesium, zinc or calcium cation.
In some preferred embodiments, the R group is methyl or ethyl, or a metal
cation
selected from a group consisting of sodium, potassium, magnesium, zinc or
calcium cation.
In an exemplary preferred embodiment, the disclosure provides compounds of the
general formula G or H, wherein: n is 9 or 11, and the fatty acid chain
contains a total of 32
or 34 carbon atoms.
In some preferred embodiments, the provided compounds G and H are
predominately one enantiomer with a defined (S) or (R) chirality at the carbon
bearing the
hydroxyl group.
In an exemplary preferred embodiment, the present disclosure provides a
compound
selected from a group consisting of I, J, K, or L, having the following
structures herein n is 9
or 11, and the fatty acid chain contains a total of 32 or 34 carbon atoms, and
the R group is
methyl or ethyl, or a metal cation selected from a group consisting of sodium,
potassium,
magnesium, or calcium cation.
(CH2)na( (CH2)n-4( (CH2)n-Y OR
OR OR
I I
=,, .. -..
I I I I I I
I HO j Ha* K HO L
0
(CH2)na(
OR
,..
I I
(R). ..... .,
HO**

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
In an exemplary preferred embodiment, the present disclosure provides compound

(S,16Z,19Z,22Z,25Z,27E,31Z)-29-hydroxytetratriaconta-16,19,22,25,27,31-
hexaenoic acid
(OR=OH), its sodium salt (OR=ONa), or its methyl ester (OR=OMe)
0
OR
HO
In other embodiments the provided compounds have the general structures of M
or
N, wherein n is a number selected from a group consisting of 0 to 19, and the
carboxylate R
group is selected from a group consisting of an ester or a pharmaceutically
acceptable salt,
wherein the R group is selected from a group consisting of hydrogen, methyl,
ethyl, alkyl, or
a cation selected from a group consisting of: ammonium cation, iminium cation,
or a metal
cation. Compounds of structure M are di-hydroxylated derivatives of very long
chain
polyunsaturated fatty acids with a total from 23 to 42 carbon atoms in the
carbon chain, two
hydroxyl groups at positions 0)-6 and o)-13, and 6 carbon-carbon double bonds
at positions
0)-3, 0)-7, 0)-9, 0)-11, 0)-15 and 0)-18. Compounds of structure N are di-
hydroxylated
derivatives of very long chain polyunsaturated fatty acids with a total from
23 to 42 carbon
atoms in the carbon chain, two hydroxyl groups at positions 0)-6 and 0)-13,
and 5 carbon-
carbon double bonds at positions co-3, co-7, co-9, ro-11 and 0)-15.
,0 ,0
OR OR
OH OH
OH OH
In preferred embodiments, n is a number selected from a group consisting of 1
to 13.
In further preferred embodiments, n is a number selected from 1, 3, 5, 7, 9,
11 or 13,
and the fatty acid contains a total of 24, 26, 28, 30, 32, 34 or 36 carbon
atoms.
In other preferred embodiments, n is a number selected from a group consisting
of 0,
2, 4, 6, 8, 10 or 12, and the fatty acid contains a total of 23, 25, 27, 19,
31, 33 or 35 carbon
atoms.
As used herein and in other structures of the present disclosure, the
compounds of
the disclosure are shown having a terminal carboxyl group "-COOR" the "R" is
intended to
designate a group covalently bonded to the carboxyl such as an alkyl group. In
the
26

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
alternative, the carboxyl group is further intended to have a negative charge
as "-coo- and
R is a cation including a metal cation, an ammonium cation and the like.
R is selected from a group consisting of methyl, ethyl, alkyl, or a cation
selected from
a group consisting of: ammonium cation, iminium cation, or a metal cation. In
some preferred
.. embodiments the metal cation is selected from a group consisting of sodium,
potassium,
magnesium, zinc or calcium cation.
In some preferred embodiments, the R group is methyl or ethyl, or a metal
cation
selected from a group consisting of sodium, potassium, magnesium, zinc or
calcium cation.
In an exemplary preferred embodiment, the disclosure provides compounds of the
general formula M or N, wherein: n is 9 or 11, and the fatty acid chain
contains a total of 32
or 34 carbon atoms.
In a preferred embodiment, the present disclosure provides a compound selected

from a group consisting of 0, P, Q, R, S, T, U or V, having the following
structures, wherein
n is 9 or 11, and the fatty acid chain contains a total of 32 or 34 carbon
atoms, and the R
group is methyl or ethyl, or a metal cation selected from a group consisting
of sodium,
potassium, magnesium, zinc or calcium cation.
,0
o
OR (CH2)n-1
OR
...OH (R) OH 01)
(s) OH
(R) OH
,0 ,0
OR OR
OH (s)'OH
(s) (R)
,0
OR (CH2)n--k
OR
OH (R) H (R)
I OH OH
(s) (R)
27

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
õ.0 0
(CH2)n¨X (C
OR 24OR
I i
==.
OH (S) .
OHH)n
U.... ".. V =,.. %%.
In an exemplary preferred embodiment, the present disclosure provides a
compound
selected from the group consisting of: (14Z,17Z,20R,21E,23E,25Z,27S,29Z)-20,27-

dihydroxydotriaconta-14,17,21,23,25,29-hexaenoic acid; sodium
(14Z,17Z,20R,21E,23E,
25Z,27S,29Z)-20,27-dihydroxydotriaconta-14,17,21,23,25,29-hexaenoate; methyl
(14Z,17Z,
20R,21E,23E,25Z,27S,29Z)-20,27-dihydroxydotriaconta-14,17,21,23,25,29-
hexaenoate;
(16Z,19Z,22R,23E,25E,27Z,29S,31Z)-22,29-dihydroxytetratriaconta-
16,19,23,25,27,31-
hexaenoic acid; sodium (16Z,19Z,22R,23E,25E,27Z,29S,31Z)-22,29-
dihydroxytetratriaconta-
16,19,23,25,27,31-hexaenoate; or methyl (16Z,19Z,22R,23E,25E,27Z,29S,31Z)-
22,29-
dihydroxy-tetratriaconta-16,19,23,25,27,31-hexaenoate, which have the
following structures:
o o o
OH ONa OMe
..
I I 1
..
OH (R) OH (R) OH (R) ...
OH
(8) 0* OH
(8) ='' OH
(e) I 1 I
-... -s. ... --.. ... -==.
0 0
0
OH
OMe
ONa
1 I 1
OH (R) ...OH OH (R)
(8)
... OH
Is)..*OH (R)
I 1 OH
(8) I
... -.. -... -.. ,.. 4%.
In other embodiments the provided compounds have the general structures of W
or
Y, wherein n is a number selected from a group consisting of 0 to 19, and the
carboxylate R
group is selected from a group consisting of an ester or a pharmaceutically
acceptable salt,
wherein the R group is selected from a group consisting of hydrogen, methyl,
ethyl, alkyl, or
a cation selected from a group consisting of: ammonium cation, iminium cation,
or a metal
cation. Compounds of structure M are di-hydroxylated derivatives of very long
chain
polyunsaturated fatty acids with a total from 23 to 42 carbon atoms in the
carbon chain, two
hydroxyl groups at positions co-6 and 0-13, and 6 carbon-carbon double bonds
at positions
co-3, co-7, co-9, co-11, co-15 and co-18. Compounds of structure N are di-
hydroxylated
derivatives of very long chain polyunsaturated fatty acids with a total from
23 to 42 carbon
28

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
atoms in the carbon chain, two hydroxyl groups at positions 0)-6 and 0)-13,
and 5 carbon-
carbon double bonds at positions 03-3, co-7, co-9, co-11 and 0)-15.
(CH2)n¨d< OR
OR
OH OH
OH OH
N-
In preferred embodiments, n is a number selected from selected from a group
consisting of 1 to 13.
In further preferred embodiments, n is a number selected from 1, 3, 5, 7, 9,
11 or 13,
and the fatty acid contains a total of 24, 26, 28, 30, 32, 34 or 36 carbon
atoms.
In other preferred embodiments, n is a number selected from a group consisting
of 0,
2, 4, 6, 8, 10 or 12, and the fatty acid contains a total of 23, 25, 27, 19,
31, 33 or 35 carbon
atoms.
R is selected from a group consisting of methyl, ethyl, alkyl, or a cation
selected from
a group consisting of: ammonium cation, iminium cation, or a metal cation. In
some preferred
embodiments the metal cation is selected from a group consisting of sodium,
potassium,
magnesium, zinc or calcium cation.
In some preferred embodiments, the R group is methyl or ethyl, or a metal
cation
selected from a group consisting of sodium, potassium, magnesium, zinc or
calcium cation.
In an exemplary preferred embodiment, the disclosure provides compounds of the

general formula W or Y, wherein: n is 9 or 11, and the fatty acid chain
contains a total of 32
or 34 carbon atoms.
In an exemplary preferred embodiment, the present disclosure provides a
compound
selected from the group consisting of compounds X or Z, wherein R is methyl or
sodium:
0
OR OR
(R)
X OH
..=OH 01)
OH
(s) (s)
ZI
Methods of preparation and manufacturing of provided compounds
The compounds provided by the present disclosure can be prepared from readily
available starting materials. For example, the synthesis of compounds of
general structure
29

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
M can be prepared according to the following general Scheme 1, which
exemplifies the
method of preparation and manufacturing of the provided compounds of this
type.
Scheme 1 shows the detailed approach for the stereocontrolled total synthesis
of
compounds of type 0, wherein n is 9, and the fatty acid chain contains a total
of 32 carbon
atoms, and the R group is methyl or sodium cation. In particular, Scheme 1
shows the
synthesis of compounds ELV1-Me and ELV1-Na, starting with methyl pentadec-14-
ynoate
(compound 4). By starting with heptadec-16-ynoate, this process affords
compounds ELV2-
Me and ELV2-Na. The alkynyl precursors of ELV1 and ELV2, namely 13a, 13b, 15a,
and
15b are also among the provided compounds X and Z in this disclosure. Scheme 1
provides
the key reagents and conditions for the preparations of the provided
compounds, by
employing reaction conditions that are typical for this type of reactions.

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Scheme 1
0 0 0
0
OMe OMe
=, Me OMe
Br
s.... N,. =
4 I cui I Nal / K2CO3 H2
__________________________ 10 I ____ li GSA
I _imp.
I
tri!i Lindlar cat.
OR1
(R) (R) (R)
OR1 (OR1 OR1
OR1 DMS0
(C001)
OR1 7 OR1 2
6 8 OH Et3N
0 0 0 0
OMe OMe OMe OMe
= = = =
Pd(PPh3)4 / Cul / Et3N Cra2ICH13 Ph3P=CHCHO
I .40t_ I welt- I i
=.-
., OR1 (R) (R)
OR1 %."' .orli
oRi po (R)
ow 0131
(S) I (3) '.. ' I o... I 6
1
,
12 11 107 9
it TBAF
0 0 0
OR OMe ONa
= = =
Zn(Cu,Ag) NaOH _low
I I I
= (R=Me) = =
ADH (R) ...OH PO
(s) I OH
(s) I J.
OH
(s) = I OH
== = ..... s...
"=,.. 13a, R=Me 14a 14b
13b, R=Na ELV1-Me ELV1-Me
0 0
OR 0
OMe
ONa
= = =
I I
= I
... OH (R) 1
(3) I OH ...OH
I(R) OH
=
..OH.s.. (R)
OH
s.. = s.. MI I
=
15a, R=Me 16a 16b
ELV2-Me ELV2-Na
15b, R=Na
Pharmaceutical compositions for the treatment of diseases
In other embodiments the present disclosure provides formulations of
pharmaceutical
compositions containing therapeutically effective amounts of one or more of
compounds
5 provided
herein or their salts thereof in a pharmaceutically acceptable carrier.
The provided compositions contain one or more compounds provided herein or
their
salts thereof, and a pharmaceutically acceptable excipient, diluent, carrier
and/or adjuvant.
The compounds are preferably formulated into suitable pharmaceutical
preparations such as
solutions, suspensions, tablets, dispersible tablets, pills, capsules,
powders, sustained
31

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
release formulations or elixirs, for oral, buccal, intranasal, vaginal,
rectal, ocular
administration or in sterile solutions or suspensions for parenteral
administration, as well as
transdermal patch preparation and dry powder inhalers. The provided
formulations may be
in the form of a drop, such as an eye drop, and the pharmaceutical formulation
may further
.. contain known agents for the treatment of eye diseases. Typically the
compounds described
above are formulated into pharmaceutical compositions using techniques and
procedures
well known in the art (see, e.g., Ansel Introduction to Pharmaceutical Dosage
Forms, Fourth
Edition 1985, 126).
Preferred embodiments of the disclosure provides pharmaceutical compositions
containing various forms of the provided compounds, as the free carboxylic
acids or their
pharmaceutically acceptable salts, or as their corresponding esters or their
phospholipid
derivatives. In other preferred embodiments the disclosure provides
pharmaceutical
compositions containing provided compounds that contain one or two hydroxyl
groups at
positions located between o-3 to o)-18 of the very long chain polyunsaturated
fatty acids, as
the free carboxylic acids or their pharmaceutically acceptable salts, or as
their corresponding
esters.
In the provided compositions, effective concentrations of one or more
compounds or
pharmaceutically acceptable derivatives is (are) mixed with a suitable
pharmaceutical carrier
or vehicle. The compounds may be derivatized as the corresponding salts,
esters, enol
ethers or esters, acids, bases, solvates, hydrates or prodrugs prior to
formulation, as
described above. The concentrations of the compounds in the compositions are
effective for
delivery of an amount, upon administration, that treats, prevents, or
ameliorates one or more
of the symptoms of a disease, disorder or condition.
As described herein, the compositions can be readily prepared by adapting
methods
known in the art. The compositions can be a component of a pharmaceutical
formulation.
The pharmaceutical formulation may further contain known agents for the
treatment of
inflammatory or degenerative diseases, including neurodegenerative diseases.
The provided
compositions can serve as pro-drug precursors of the fatty acids and can be
converted to
the free fatty acids upon localization to the site of the disease.
The present disclosure also provides packaged composition(s) or pharmaceutical
composition(s) for use in treating the disease or condition. Other packaged
compositions or
pharmaceutical compositions provided by the present disclosure further include
indicia
including at least one of: instructions for using the composition to treat the
disease or
condition. The kit can further include appropriate buffers and reagents known
in the art for
administering various combinations of the components listed above to the host.
Pharmaceutical formulations
32

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Embodiments of the present disclosure include a composition or pharmaceutical
composition as identified herein and can be formulated with one or more
pharmaceutically
acceptable excipients, diluents, carriers and/or adjuvants. In addition,
embodiments of the
present disclosure include a composition or pharmaceutical composition
formulated with one
or more pharmaceutically acceptable auxiliary substances. In particular the
composition or
pharmaceutical composition can be formulated with one or more pharmaceutically

acceptable excipients, diluents, carriers, and/or adjuvants to provide an
embodiment of a
composition of the present disclosure.
A wide variety of pharmaceutically acceptable excipients are known in the art.
Pharmaceutically acceptable excipients have been amply described in a variety
of
publications, including, for example, A. Gennaro (2000) "Remington: The
Science and
Practice of Pharmacy," 20th edition, Lippincott, Williams, 8, Wilkins;
Pharmaceutical Dosage
Forms and Drug Delivery Systems (1999) H.C. Ansel et al., eds., 7th ed.,
Lippincott,
Williams, 8, Wilkins; and Handbook of Pharmaceutical Excipients (2000) A.H.
Kibbe et al.,
eds., 3rd ed. Amer. Pharmaceutical Assoc. The pharmaceutically acceptable
excipients,
such as vehicles, adjuvants, carriers or diluents, are readily available to
the public.
Moreover, pharmaceutically acceptable auxiliary substances, such as pH
adjusting and
buffering agents, tonicity adjusting agents, stabilizers, wetting agents and
the like, are readily
available to the public.
In an embodiment of the present disclosure, the composition or pharmaceutical
composition can be administered to the subject using any means capable of
resulting in the
desired effect. Thus, the composition or pharmaceutical composition can be
incorporated
into a variety of formulations for therapeutic administration. For example,
the composition or
pharmaceutical composition can be formulated into pharmaceutical compositions
by
combination with appropriate, pharmaceutically acceptable carriers or
diluents, and may be
formulated into preparations in solid, semi-solid, liquid or gaseous forms,
such as tablets,
capsules, powders, granules, ointments, solutions, suppositories, injections,
inhalants and
aerosols.
Suitable excipient vehicles for the composition or pharmaceutical composition
are, for
example, water, saline, dextrose, glycerol, ethanol, or the like, and
combinations thereof. In
addition, if desired, the vehicle may contain minor amounts of auxiliary
substances such as
wetting or emulsifying agents or pH buffering agents. Methods of preparing
such dosage
forms are known, or will be apparent upon consideration of this disclosure, to
those skilled in
the art. See, e.g., Remington's Pharmaceutical Sciences, Mack Publishing
Company,
Easton, Pennsylvania, 17th edition, 1985. The composition or formulation to be
administered
will, in any event, contain a quantity of the composition or pharmaceutical
composition
adequate to achieve the desired state in the subject being treated.
33

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Compositions of the present disclosure can include those that comprise a
sustained
release or controlled release matrix. In addition, embodiments of the present
disclosure can
be used in conjunction with other treatments that use sustained-release
formulations. As
used herein, a sustained-release matrix is a matrix made of materials, usually
polymers,
which are degradable by enzymatic or acid-based hydrolysis or by dissolution.
Once inserted
into the body, the matrix is acted upon by enzymes and body fluids. A
sustained-release
matrix desirably is chosen from biocompatible materials such as liposomes,
polylactides
(polylactic acid), polyglycolide (polymer of glycolic acid), polylactide co-
glycolide (copolymers
of lactic acid and glycolic acid), polyanhydrides, poly(ortho)esters,
polypeptides, hyaluronic
acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids,
phospholipids,
polysaccharides, nucleic acids, polyamino acids, amino acids such as
phenylalanine,
tyrosine, isoleucine, polynucleotides, polyvinyl propylene,
polyvinylpyrrolidone and silicone.
Illustrative biodegradable matrices include a polylactide matrix, a
polyglycolide matrix, and a
polylactide co-glycolide (co-polymers of lactic acid and glycolic acid)
matrix. In another
embodiment, the pharmaceutical composition of the present disclosure (as well
as
combination compositions) can be delivered in a controlled release system. For
example, the
composition or pharmaceutical composition may be administered using
intravenous infusion,
an implantable osmotic pump, a transdermal patch, liposomes, or other modes of

administration. In one embodiment, a pump may be used (Sefton (1987). CRC
Crit. Ref.
Biomed. Eng. 14:201; Buchwald et al. (1980). Surgery 88:507; Saudek et al.
(1989). N. Engl.
J. Med. 321:574). In another embodiment, polymeric materials are used. In yet
another
embodiment a controlled release system is placed in proximity of the
therapeutic target thus
requiring only a fraction of the systemic dose. In yet another embodiment, a
controlled
release system is placed in proximity of the therapeutic target, thus
requiring only a fraction
of the systemic. Other controlled release systems are discussed in the review
by Langer
(1990). Science 249:1527-1533.
In another embodiment, the compositions of the present disclosure (as well as
combination compositions separately or together) include those formed by
impregnation of
the composition or pharmaceutical composition described herein into absorptive
materials,
such as sutures, bandages, and gauze, or coated onto the surface of solid
phase materials,
such as surgical staples, zippers and catheters to deliver the compositions.
Other delivery
systems of this type will be readily apparent to those skilled in the art in
view of the instant
disclosure.
In another embodiment, the compositions or pharmaceutical compositions of the
present disclosure (as well as combination compositions separately or
together) can be part
of a delayed-release formulation. Delayed-release dosage formulations can be
prepared as
described in standard references such as "Pharmaceutical dosage form tablets",
eds.
34

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Liberman et. al. (New York, Marcel Dekker, Inc., 1989), "Remington ¨The
science and
practice of pharmacy", 20th ed., Lippincott Williams 8, Wilkins, Baltimore,
MD, 2000, and
"Pharmaceutical dosage forms and drug delivery systems", 6th Edition, Ansel et
al., (Media,
PA: Williams and Wilkins, 1995). These references provide information on
excipients,
.. materials, equipment and process for preparing tablets and capsules and
delayed release
dosage forms of tablets, capsules, and granules. These references provide
information on
carriers, materials, equipment and process for preparing tablets and capsules
and delayed
release dosage forms of tablets, capsules, and granules.
Embodiments of the composition or pharmaceutical composition can be
administered
to a subject in one or more doses. Those of skill will readily appreciate that
dose levels can
vary as a function of the specific the composition or pharmaceutical
composition
administered, the severity of the symptoms and the susceptibility of the
subject to side
effects. Preferred dosages for a given compound are readily determinable by
those of skill in
the art by a variety of means.
In an embodiment, multiple doses of the composition or pharmaceutical
composition
are administered. The frequency of administration of the composition or
pharmaceutical
composition can vary depending on any of a variety of factors, e.g., severity
of the
symptoms, and the like. For example, in an embodiment, the composition or
pharmaceutical
composition can be administered once per month, twice per month, three times
per month,
every other week (qow), once per week (qw), twice per week (biw), three times
per week
(tiw), four times per week, five times per week, six times per week, every
other day (qod),
daily (qd), twice a day (qid), three times a day (tid), or four times a day.
As discussed above,
in an embodiment, the composition or pharmaceutical composition is
administered 1 to 4
times a day over a 1 to 10 day time period.
The duration of administration of the composition or pharmaceutical
composition
analogue, e.g., the period of time over which the composition or
pharmaceutical composition
is administered, can vary, depending on any of a variety of factors, e.g.,
patient response,
etc. For example, the composition or pharmaceutical composition in combination
or
separately, can be administered over a period of time of about one day to one
week, about
one day to two weeks.
The amount of the compositions and pharmaceutical compositions of the present
disclosure that can be effective in treating the condition or disease can be
determined by
standard clinical techniques. In addition, in vitro or in vivo assays can
optionally be employed
to help identify optimal dosage ranges. The precise dose to be employed can
also depend
on the route of administration, and can be decided according to the judgment
of the
practitioner and each patient's circumstances.
Routes of Administration

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Embodiments of the present disclosure provide methods and compositions for the

administration of the active agent(s) to a subject (e.g., a human) using any
available method
and route suitable for drug delivery, including in vivo and ex vivo methods,
as well as
systemic and localized routes of administration. Routes of administration
include intranasal,
.. intramuscular, intratracheal, subcutaneous, intradermal, topical
application, intravenous,
rectal, nasal, oral, and other enteral and parenteral routes of
administration. Routes of
administration may be combined, if desired, or adjusted depending upon the
agent and/or
the desired effect. An active agent can be administered in a single dose or in
multiple doses.
The VLC-PUFA and their biogenic derivatives are formed in cells and are not a
component of human diet. Possible routes of administration of the novel
compounds
provided herein will include oral and parenteral administration, including
intravitreal and
subretinal injection into the eye to by-pass intestinal absorption, the gut-
liver, and the blood¨
ocular barrier. The provided formulations may be delivered in the form of a
drop, such as an
eye drop, or any other customary method for the treatment of eye diseases.
Parenteral routes of administration other than inhalation administration
include, but
are not limited to, topical, transdermal, subcutaneous, intramuscular,
intraorbital,
intracapsular, intraspinal, intrasternal, and intravenous routes, i.e., any
route of
administration other than through the alimentary canal. Parenteral
administration can be
conducted to effect systemic or local delivery of the composition. Where
systemic delivery is
desired, administration typically involves invasive or systemically absorbed
topical or
mucosal administration of pharmaceutical preparations. In an embodiment, the
composition
or pharmaceutical composition can also be delivered to the subject by enteral
administration.
Enteral routes of administration include, but are not limited to, oral and
rectal (e.g., using a
suppository) delivery.
Methods of administration of the composition or pharmaceutical composition
through
the skin or mucosa include, but are not limited to, topical application of a
suitable
pharmaceutical preparation, transdermal transmission, injection and epidermal
administration. For transdermal transmission, absorption promoters or
iontophoresis are
suitable methods. lontophoretic transmission may be accomplished using
commercially
available "patches" that deliver their product continuously via electric
pulses through
unbroken skin for periods of several days or more.
Methods for the treatment of diseases, disorders or conditions
Described herein are methods and compositions for treating and protecting an
organ
or tissue from the effects of oxidative stress or other homeostatic
disruptions associated with
a persistent inflammatory condition or a progressive degenerative disease,
including a
neurodegenerative disease.
36

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
The provided compounds, compositions, and methods can be used for the
preventive
and therapeutic treatment of a disease, disorder or condition. The list of
diseases that can be
treated with the provided compositions and methods include but are not limited
to
inflammatory diseases, degenerative diseases, including neurodegenerative
diseases
including, but not limited to the following:
(a) Inflammatory diseases, including acute and chronic disorders were
homeostasis
is disrupted by abnormal or dysregulated inflammatory response. These diseases
are
initiated and mediated by a number of inflammatory factors, including
oxidative stress,
chemokines, cytokines, breakage of blood/tissue barriers, autoimmune diseases
or other
conditions that engage leukocytes, monocytes/macrophages or parenchymal cells
that
induce excessive amounts of pro-cell injury, pro-inflammatory/disruptors of
homeostasis
mediators. These diseases occur in a wide range of tissues and organs and are
currently
treated, by anti-inflammatory agents such as corticosteroids, non-steroidal
anti-inflammatory
drugs, TNF modulators, COX-2 inhibitors, etc. Representative examples include
but are not
limited to: rheumatoid arthritis, osteoarthritis, atherosclerosis, cancer,
diabetes, intestinal
bowel disease, prostatitis, ischemic stroke, traumatic brain damage, spinal
cord injury,
multiple sclerosis, autism, schizophrenia, depression, traumatic brain injury,
status
epilepticus, Huntington's disease, Alzheimer's disease, Parkinson's disease,
amyotrophic
lateral sclerosis, retina degenerative diseases, age-related macular
degeneration, inherited
retinal degenerative diseases, Stargardt-like macular dystrophy, X-linked
juvenile
retinoschisis, perioperative hypoxia, retinitis pigmentosa, glaucoma, etc.
(b) Degenerative diseases, which include conditions that involve progressive
loss of
vital cells and tissues that result in progressive impairment of function,
such as loss of
cartilage in knees, hip joints or other joints such as in osteoarthritis.
Other degenerative
diseases engages cellular and intercellular homeostasis perturbations and
includes heart
disease, atherosclerosis, cancer, diabetes, intestinal bowel disease,
osteoporosis,
prostatitis, rheumatoid arthritis, etc.
(c) Neurodegenerative diseases, which include some of the major diseases of
the
brain, retina, spinal cord and peripheral nerves, whereby a progressive demise
of cellular
organization leads to impaired function. These are due to immune or
inflammatory disorders
and/or to inherited conditions or aging. They include ischemic stroke,
traumatic brain
damage, spinal cord injury, epilepsy, multiple sclerosis, Alzheimer's disease,
Parkinson's
disease, amyotrophic lateral sclerosis, retina degenerative diseases such as
age-related
macular degeneration, inherited eye diseases such as retinitis pigmentosa,
glaucoma, etc.
(d) Retinal degenerative diseases, which are the leading causes of blindness
that
affects very large numbers of people and involve the deterioration of the
retina caused by
the progressive and eventual death of the photoreceptor cells of the retina.
Examples of
37

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
common retinal degenerative diseases include but are not limited to: retinitis
pigmentosa,
age-related macular degeneration, inherited retinal degenerative diseases,
Stargardt-like
macular dystrophy, X-linked juvenile retinoschisis, perioperative hypoxia,
glaucoma, etc.
The provided compounds, compositions, and methods can also be used to induce
the increased expression of Sirtuin1 (SIRT1) and to treat diseases and
conditions that can
benefit from an increased expression of SI RT1. Sirtuin1 (SI RT1) belongs to a
family of
highly conserved proteins associated with aging, modulation of energy
metabolism, genomic
stability, stress resistance, Alzheimer's and other neurodegenerative
diseases. Sirtuin1 is a
major therapeutic target in many diseases including cancer, diabetes,
inflammatory
disorders and neurodegenerative disease, all of which can be treated with the
provided
compounds, compositions and methods.
Also described herein are methods and compositions for treating and protecting
the
retina of the eye. Specifically, described herein are methods for treating and
protecting
retinal pigment epithelial cells and photoreceptors of the eye. Generally,
compositions as
described herein are administered to a subject in any preferred mode of
administration. Such
modes include in an eye drop.
Methods and compositions described herein can be used to treat a diseased eye
in a
subject. For example, the disease can be a retinal disease, such as retinal
degeneration. In
this instance, the retinal degeneration can be prevented or delayed. Eye
diseases that are
particularly suited for methods and compositions as described herein include
age-related
macular degeneration, retinitis pigmentosa, and Stargardt disease.
Methods and compositions described herein can promote the survival of
photoreceptors in the retina.
Methods and compositions described herein can induce signaling pathways that
enhance cell survival in cell specific to the eye, such as retinal pigment
epithelial cells and
photoreceptors.
While embodiments of the present disclosure are described in connection with
the
Examples and the corresponding text and figures, there is no intent to limit
the disclosure to
the embodiments in these descriptions. On the contrary, the intent is to cover
all alternatives,
modifications, and equivalents included within the spirit and scope of
embodiments of the
present disclosure.
EXAMPLES
Example 1
Evidence for the formation of hydroxylated VLC-PUFA in cells as postulated in
Figure
1, and as documented in Figure 2. The biogenic conversion of the n3 VLC-PUFA
(1) to the
mono-hydroxylated derivative 2 and the di-hydroxylated derivative 3
demonstrates the ability
of VLC-PUFA to generate hydroxylated derivatives with structures analogous to
those
38

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
obtained from other omega-3 PUFA such as DHA. The preferred structures of 2
are 2a and
2b, while the preferred structures of 3 are 3a and 3b. Although these novel
findings do not
prove that compounds 2 or 3 are naturally occurring in living systems, they
provide a design
rationale for the provided compounds and their biological activities, as
provided in this
disclosure. The cell-derived hydroxylated derivatives 2 and 3 were obtained
from human
retinal pigment epithelial cells in culture. Human retinal pigment epithelial
cells
(spontaneously transformed ARPE-19 cells) or primary human retinal pigment
epithelial cells
(HRPE) were incubated with 34:6n3 (100nM) during 12-16 hours and then the
culture media
collected ,lipid extracted and run in LC-MS/MS. The results suggest that C34:6
with an m/z
of 495.5 (Figure 2A) yielded a hydroxylated product analogous to the mono-
hydroxylated
DHA derivative 17-HDHA having a parent-H m/z of 511.8 and a fragment m/z of
413, which
is consistent with the mono-hydroxylated compound 29-hydroxy-34:6 (Figure 2B).
The data
show that compound C34:6 was also converted to an elongation product analogous
to the
di-hydroxylated DHA derivative NPD1,(10R,17S-dihydroxy-docosa-
4Z,7Z,11E,13E,15Z,19Z-
hexaenoic acid) having a parent-H m/z of 527.8, and a fragment m/z of 206
consistent with
the NPD1-like di-hydroxylated compound 22,29-dihydroxy-34:6 (Figure 2C). The
stereochemistry of the mono- and di-hydroxy compounds were not possible to
determine,
but they were presumed to be the same as those derived from DHA.
,o 1 ,s3
o
(CH2),-46 (CH On ¨4(
OH
I 1
', =.,
(cHon¨xD 1 1 1 1 1 I
OH
HO HO HO
=..
2
/9 2a
/2 2b
,9
OH OH
OH
.... .00 =-=-õ,,_116.
.., =. ".
1
I I I
n3 VLC-PUFA *. =.,
OH OH (R) OH (11)
1 OH .,
OH
OH
09 I (11) I
3
3a 3b
Example 2
Representative of experiments used human retinal pigment epithelial (RPE)
cells,
which are neuroectoderm-derived post-mitotic cells of the retina, an integral
part of the
central nervous system. These cells are richly endowed with a multitude of
mechanisms to
protect themselves from injury and to protect other cells, particularly the
survival of
photoreceptors. They are the most active phagocyte of the human body, critical
for the
39

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
health of photoreceptors and vision, and have the ability to secrete
neurotrophins and other
beneficial substances. In pathological conditions they recapitulate aspects of
Alzheimer's
disease by processing amyloid precursor protein and contributing to the
formation of Drusen,
analogously to the senile amyloid plaques. Thus, these are among the reasons
that
experimental data included in this disclosure were obtained with RPE cells.
Therefore, the
data provided herein are representative of the expected activities of the
provided compounds
in other cells and tissues where VLC-PUFA are known to be generated or be
present.
Evidence of cytoprotection by 32.6 and 34.6 VLC-PUFA in oxidative-stress
induced
ARPE-19 cells as detailed in (Figure 4): (A) Cytoprotective effects of NPD1
like compounds
on OS induced apoptosis. The results shown in this figure compare the
cytoprotective
capacities of very long chain polyunsaturated fatty acids (VLC-PUFA),
elovanoids ELV1 and
ELV2, and neuroprotectin D1 (NPD1) in human retinal pigment epithelial (RPE)
cells
deficient in 15-LOX-D1 by measuring the protection of cell deaths induced by
oxidative
stress (OS) by these compounds. The results indicate that NPD1 provided the
maximum
protection (60%), followed by elovanoids at intermediate level (55%), and VLC-
PUFA (50%),
the least compared to OS (90%). (B) Elovanoid precursors protect human retinal
pigment
epithelial cells deficient in 15-LOX-1, unlike DHA, from oxidative stress
conditions. This
experiment clearly shows that VLC-PUFA, elovanoid precursors 32:6 and 34:6,
and NPD1
protect against cell death in 15-LOX-D1 cells under oxidative stress
conditions. On the other
hand, DHA was unable to do so, as the 15-LOX-D1 cells lack the enzyme required
for
conversion of DHA to the neuroprotective agent.
Example 3
Figure 5- (A) Effect of PD146176 on VLC-PUFA inhibited apoptosis induced by OS
in ARPE-19 cells. This experiment demonstrated the effect of 15-lipooxygenese
inhibitor
PD146176 on the VLC-PUFA-mediated inhibition of cell death in ARPE-19 cells
under
stressed condition. It is evident that 32:6 and 34:6 LCAF were able to induce
a substantial
amount (55 and 48% respectively) of neuroprotection compared to PD 146176
(22%) when
the stressed cells were treated with 5pm of 15-LOX-D1 inhibitor. It can be
concluded that
since PD146176 is the inhibitor of 15-lipooxygenase enzyme, it might be
protecting the
stressed RPE cells by accumulating neuroprotective agents inside the cells.
(B) Comparison
of cytoprotective capacities of NPD1, C32:6 and C34:6 VLC-PUFA on oxidative
stress-
induced apoptosis in 15-LOX-1 cells. Shown here is the comparison of
neuroprotection in a
15-LOX-1-deficient cell line under oxidative stress with 32:6 and 34:6 VLC-
PUFA along with
NPD1. 32:6 and 34:6 VLC-PUFA were able to induce neuroprotection (45% and 40%
respectively), as compared to oxidative stress (90%) under this condition. (C)
Concentration
dependent cytoprotection by C32:6 and C34:6 VLC-PUFA in oxidative-stress
induced ARPE-
19 cells. A concentration (50-500nM) kinetic of cytoprotection induced by VLC-
PUFA 32:6

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
and 34:6 in RPE cell culture under OS was shown here. The result indicates
that there was a
gradual decrease of cell deaths starting from 50nM concentrations of both 32:6
and 34:6
VLC-PUFA, very good intermediate effect at 250nM, and maximum effect at 500nM.
We
decided to use 250nM concentrations of 32:6 and 34:6 VLC-PUFA in subsequent
experiments. (D) Selected images of alive and dead cells from this study
(control, OS,
treatment with C32:6).
Example 4
Figure 6- (A) VLC-PUFA and elovanoids ELV1 and ELV2 mediated effect on Bid
upregulation in ARPE-19 cells under stress. This figure displays the
downregulation of the
proapoptotic protein of the BcI2 family Bid by western blot analysis by VLC-
PUFA and
elovanoids in RPE cells in culture under oxidative-stress. Results indicate
that upregulated
Bid protein by OS, as evident from the figure, was inhibited by both
elovanoids and VLC-
PUFA. It is interesting to see that the sodium salts of the elovaniod
precursors are more
effective than the methyl ester forms. (B) VLC-PUFA and ELV1 and ELV2
compounds
mediated upregulation of Bid in ARPE-19 cells under stress. This Figure shows
the
quantification of Bid downregulation.
Example 5
Figure 7- A) VLC-PUFA and ELV1 and ELV2 compounds mediated upregulation of
Bim in ARPE-19 cells under stress. Bim, another class of BcI2 family, has been
tested like
Bid in this figure to confirm our previous results. VLC-PUFA and elovanoids
protected the
upregulation of Bim by OS, similar to Bid, in RPE cells under stress. (B) VLC-
PUFA and
elovanoids mediated effect on Bim upregulation in ARPE-19 cells under stress.
This Figure
shows the quantification of Bim downregulation.
Example 6
Figure 8- (A) Bc1-xL-upregulation by elovanoids ELV1 and ELV2 in ARPE-19 cells
under stress. BcI-xL is the antiapoptotic BcI2 family protein. Like
proapoptotic proteins Bid
and Bim, the effect of elovaniod precursors on the antiapoptotic protein BcI-
xL was tested in
this figure in RPE cells under OS. Results showed that elovaniod precursors
were able to
upregulate the BcI-xL protein in RPE cells under stress, which is the opposite
effect of Bid
and Bim. (B) Effect of NPD1, ELV1 and ELV2 on Bax expression in LOX-D cells
under
stress. Proapoptotic Bax was tested in this figure. It is evident that
elovaniod precursors
downregulated the Bax upregulation by OS in RPE cells under OS, which is
consistent with
our inhibition of apoptosis experiments, as shown before. C) VLC-PUFA and
elovanoids
ELV1 and ELV2 mediated effect on Bax upregulation in ARPE-19 cells under
stress. In this
experiment, elovanoid precursors along with VLC-PUFA were tested on the
downregulation
of the Bax protein in RPE cells under stress.
Example 7
41

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Figure 9- (A) VLC-PUFA and elovanoids ELV1 and ELV2 mediated effect on BcI2
upregulation in ARPE-19 cells under stress. In this experiment we tested the
effect of
elovaniod precursors on B1c2 upregulation along with VLC-PUFA in stressed RPE.
(B)
Quantification of BcI2 upregulation by NPD1, ELV1 and ELV2 in LOX-D cells.
BcI2 is an
important antiapoptotic protein of the BcI2 family protein. It is evident that
elovaniod
precursors upregulated the BcI2 protein in RPE cells under stress.
Example 8
Figure 10 - (A) Effect of NPD1 and VLC-PUFA C32:6 and C34:6 in mediating
upregulation of SIRT1 in ARPE-19 cells. (B) Quantification of SIRT1
upregulation by NPD1,
C32:6 and C34:6. SIRT1 (Sirtuin1) belongs to a family of highly conserved
proteins linked to
caloric restriction beneficial outcomes and aging by regulating energy
metabolism, genomic
stability and stress resistance. SIRT1 is a potential therapeutic target in
several diseases
including cancer, diabetes, inflammatory disorders, and neurodegenerative
diseases or
disorders. Elovanoids induce cell survival involving the upregulation of the
BcI2 class of
survival proteins and the downregulation of pro-apoptotic Bad and Bax under
oxidative
stress (OS) in RPE cells. The data in this Figure suggest that elovanoids
upregulate SI RT1
abundance in human RPE cells when confronted with OS. As a consequence,
remarkable
cell survival takes place. This target of elovanoids might be relevant to
counteract
consequences of several diseases associated with SIRT1.
Example 9
Figure 11 - The elovanoid ELV2 in 200 nM concentrations protects neuronal
cells in
primary cultures from NMDA-induced toxicity (A), and MK-801 potentiates
protection as
assessed by MTT assay for cell viability (B). In several neurological and
neurodegenerative
diseases, such as stroke, epilepsy, status epilepticus, traumatic head injury,
etc., as well as
ophthalmological diseases, such as glaucoma, an excessive presynaptic release
of the
excitatory neurotransmitter glutamate takes place. As a consequence, glutamate

transporters that function to remove extracellular glutamate from astrocytes
and neurons are
overwhelmed and the NMDA-type glutamate receptor is over-activated. This
receptor is a
calcium channel that therefore leads to a flooding of calcium into the
postsynaptic cell. The
overall phenomena is refer to as excitotoxicity that in turn leads to neuronal
damage and cell
death. MK801 is a known blocker of this receptor used here as a control. The
results in this
Figure demonstrate that when NMDA in increasing concentrations is added to
neuronal
cultures it leads to cell death, while the use of ELV2 reduces cell death and
increases cell
viability. These data support the use of the elovanoids for the treatment of
neurodegenerative diseases and conditions involving NMDA-related
excitotoxicity, such as:
ischemic stroke, Alzheimer's disease, Parkinson's disease, etc.
Example 10
42

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Figure 12 - Elovanoids ELV2-Na and ELV2-Me are more active than DHA and NPD1
in a model of ischemic stroke after 2hrs of MCAo (middle cerebral occlusion).
To test the
novel elovanoids the experimental design consisted injecting the compounds
into the right
cerebral ventricle (5 pg/per rat, ICV), one hour after two hours of an
ischemic stroke in rats
and following thereafter the neurological behavior (neurological score) during
7 days. The
protocol in brief was as follows. The injection was made through a surgically
implanted metal
canula (Alzet) into the right lateral ventricle. Two days later the right
middle cerebral artery
(MCA) was occluded for 2 h by means of an intraluminal nylon filament (Belayev
et al,
Traslational Stroke Research, 2010). Then one hour after the compounds were
injected
dissolved in sterile cerebrospinal fluid. The occlusion was transient
performed as follows.
The right common carotid artery was exposed through an incision in the neck
and was
isolated from surrounding tissues. The distal external carotid artery and
pterygopalatine
arteries were tied. A 4-cm of 3-0 monofilament nylon suture coated with poly-
Lysine was
introduced into the internal carotid artery and MCA. The suture position was
confirmed by
advancing the suture 20-22 mm from the common carotid artery bifurcation.
Then, the rats
were allowed to awaken from anesthesia and returned to their cages. The degree
of stroke
injury was assessed by neurological assessment of each rat at 60 min after
onset of MCAo.
Rats that do not demonstrate high-grade contralateral deficit (score, 10-11)
were excluded
from the study. After 2 hours of MCA occlusion, the rats were re-anesthetized
temperature
probes were reinserted and the intraluminal suture was removed. The neck
incision was
closed with silk sutures, and the animals were allowed free access to food and
water. These
results show that the use elovanoids after the ischemic event result in
remarkable
neuroprotection, suggesting a potential therapeutic benefit for the treatment
of ischemic
stroke and other neurodegenerative diseases or disorders. The sodium salt ELV2-
Na
showed a greater potency, presumably because it delivers the active form of
ELV2 and has
a more immediate effect. The methyl ester ELV2-Me is expected to first be
hydrolyzed via
the actions of esterases, and it may have a more delayed effect, which may be
beneficial for
a sustainable long-term treatment. Overall, these data demonstrate the
neuroprotective
effects of the elovanoids, either as pharmaceutically acceptable salts (e.g.
ELV2-Na), or in
the form of a prodrug, such as an ester derivative (e.g. ELV2-Me), or as a
pharmaceutical
composition containing a combination of the two forms that can have both an
immediate and
a sustainable long-term therapeutic effect.
REFERENCES
Bazan NG. Neuroprotectin D1 (NPD1): A DHA-derived mediator that protects brain
and
retina against cell injury-induced oxidative stress. Brain Pathology.
2005;15(2):159-66.
Bazan NG, Eady TN, Khoutorova L, Atkins KD, Hong S, Lu Y, Zhang C, Jun B,
Obenaus A,
Fredman G, Zhu M, Winkler JW, Petasis NA, Serhan CN, Belayev L. Novel aspirin-
triggered
43

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
neuroprotectin D1 attenuates cerebral ischemic injury after experimental
stroke. Exp Neural.
2012; 236(1):122-30.
Bazan NG, Molina MF, Gordon WC. Docosahexaenoic acid signalolipidomics in
nutrition:
Significance in aging, neuroinflammation, macular degeneration, Alzheimer's,
and other
neurodegenerative diseases. Annual Review of Nutrition, 2011;31(1):321-51.
Bennett LD, Brush RS, Chan M, Lydic TA, Reese K, Reid GE, Busik JV, Elliott
MH,
Anderson RE. Effect of Reduced Retinal VLC-PUFA on Rod and Cone
PhotoreceptorsRetinal VLC-PUFA in Rod and Cone Photoreceptors. Investigative
Ophthalmology & Visual Science. 2014;55(5):3150-7.
Brush RS, Tran J-TA, Henry KR, McClellan ME, Elliott MH, Mandal MNA. Retinal
Sphingolipids and Their Very-Long-Chain Fatty Acid¨Containing Species.
Investigative
Ophthalmology & Visual Science. 2010;51(9):4422-31.
Harkewicz R, Du H, Tong Z, Alkuraya H, Bedell M, Sun W, Wang X, Hsu Y-H,
Esteve-Rudd
J, Hughes G, Su Z, Zhang M, Lopes VS, MoIday RS, Williams DS, Dennis EA, Zhang
K.
Essential Role of ELOVL4 Protein in Very Long Chain Fatty Acid Synthesis and
Retinal
Function. J Biol Chem. 2012;287(14):11469-80.
Kihara A. Very long-chain fatty acids: elongation, physiology and related
disorders. Journal
of Biochemistry. 2012;152(5):387-95.
Liu A, Lin Y, Terry R, Nelson K, Bernstein PS. Role of long-chain and very-
long-chain
polyunsaturated fatty acids in macular degenerations and dystrophies. Clinical
Lipidology.
2011;6(5):593-613.
Logan S, Agbaga M-P, Chan MD, Kabir N, Mandal NA, Brush RS, Anderson RE.
Deciphering mutant ELOVL4 activity in autosomal-dominant Stargardt macular
dystrophy.
Proc Natl Acad Sci USA. 2013;110(14):5446-51.
.. Mattson MP, Bazan NG: Apoptosis and necrosis. In Basic Neurochemistry, 7th
edition,
G.Siegel, R.W. Albers, S.T. Brady, D.L. Price, eds, 2005.
McMahon A, Jackson SN, Woods AS, Kedzierski W. A Stargardt disease-3 mutation
in the
mouse ElovI4 gene causes retinal deficiency of C32¨C36 acyl
phosphatidylcholines. FEBS
Letters. 2007;581(28):5459-63
Okuda A, Naganuma T, Ohno Y, Abe K, Yamagata M, lgarashi Y, Kihara A. Hetero-
oligomeric interactions of an ELOVL4 mutant protein: implications in the
molecular
mechanism of Stargardt-3 macular dystrophy. J Molecular Vision. 2010;16:2438-
45.
Olson DK, Frahlich F, Christiana R, Hannibal-Bach HK, Ejsing CS, Walther TC.
Rom2-
dependent Phosphorylation of Elo2 Controls the Abundance of Very Long-chain
Fatty Acids.
Journal of Biological Chemistry. 2015;290(7):4238-47.
Petasis NA, Yang R, Winkler JW, Zhu M, Uddin J, Bazan NG, Serhan CN.
Stereacontrolled
total synthesis of Neuroprotectin D1/Protectin D1 and its aspirin-triggered
stereoisomer.
44

CA 03014033 2018-08-08
WO 2016/130522
PCT/US2016/017112
Tetrahedron Left. 2012;53(14):1695-8.
Poulos A, Johnson DW, Beckman K, White IG, Easton C. Occurrence of unusual
molecular
species of sphingomyelin containing 28-34-carbon polyenoic fatty acids in ram
spermatozoa.
Biochemical Journal. 1987;248(3):961-4.
Rice DS, Calandria JM, Gordon WC, Jun B, Zhou Y, Gelfman CM, Li S, Jin M,
Knott EJ,
Chang B, Abuin A, lssa T, Potter D, Platt KA, Bazan NG. Adiponectin receptor 1
conserves
docosahexaenoic acid and promotes photoreceptor cell survival. Nat Commun.
2015;6:1-14.
doi: 10.1038/ncomms7228.
Serhan CN, Gotlinger K, Hong S, Lu Y, Siegelman J, Baer T, Yang R, Colgan SP,
Petasis
NA. Anti-inflammatory actions of neuroprotectin D1/protectin D1 and its
natural
stereoisomers: Assignments of dihydroxy-containing docosatrienes. J. lmmunol.
2006;176(3):1848-59.
Serhan CN, Petasis NA. Resolvins and protectins in inflammation resolution.
Chem Rev.
2011;111(10):5922-43.
Stark DT, Bazan NG. Synaptic and Extrasynaptic NMDA Receptors Differentially
Modulate
Neuronal Cyclooxygenase-2 Function, Lipid Peroxidation, and Neuroprotection.
The Journal
of Neuroscience. 2011;31(39):13710-21.
Zhou Q, Sheng M. NMDA receptors in nervous system diseases. Neuropharmacology.

2013;74:69-75.
45

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2024-02-27
(86) PCT Filing Date 2016-02-09
(87) PCT Publication Date 2016-08-18
(85) National Entry 2018-08-08
Examination Requested 2021-01-14
(45) Issued 2024-02-27

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $277.00 was received on 2024-02-02


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2025-02-10 $100.00
Next Payment if standard fee 2025-02-10 $277.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Reinstatement of rights $200.00 2018-08-08
Application Fee $400.00 2018-08-08
Maintenance Fee - Application - New Act 2 2018-02-09 $100.00 2018-08-08
Maintenance Fee - Application - New Act 3 2019-02-11 $100.00 2018-08-08
Maintenance Fee - Application - New Act 4 2020-02-10 $100.00 2020-01-31
Request for Examination 2021-02-09 $816.00 2021-01-14
Maintenance Fee - Application - New Act 5 2021-02-09 $204.00 2021-02-05
Maintenance Fee - Application - New Act 6 2022-02-09 $203.59 2022-02-04
Maintenance Fee - Application - New Act 7 2023-02-09 $210.51 2023-02-03
Final Fee $416.00 2024-01-19
Maintenance Fee - Application - New Act 8 2024-02-09 $277.00 2024-02-02
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
UNIVERSITY OF SOUTHERN CALIFORNIA
BOARD OF SUPERVISORS OF LOUISIANA STATE UNIVERSITY AND AGRICULTURAL AND MECHANICAL COLLEGE
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Request for Examination 2021-01-14 4 146
Examiner Requisition 2022-03-24 4 237
Amendment 2022-07-22 30 1,137
Description 2022-07-22 45 3,543
Claims 2022-07-22 9 324
Amendment 2022-10-17 4 102
Examiner Requisition 2022-12-09 3 186
Amendment 2023-04-11 25 724
Claims 2023-04-11 10 346
Electronic Grant Certificate 2024-02-27 1 2,527
Abstract 2018-08-08 2 76
Claims 2018-08-08 9 242
Drawings 2018-08-08 13 638
Description 2018-08-08 45 2,551
Representative Drawing 2018-08-08 1 16
International Preliminary Report Received 2018-08-08 11 713
International Search Report 2018-08-08 3 120
Declaration 2018-08-08 2 67
National Entry Request 2018-08-08 2 111
Cover Page 2018-08-16 1 45
Final Fee 2024-01-19 5 159
Representative Drawing 2024-01-31 1 10
Cover Page 2024-01-31 1 46
Interview Record Registered (Action) 2023-08-02 1 25
Amendment 2023-09-05 25 693
Claims 2023-09-05 10 349